1
|
Greiner J, Dente M, Orós-Rodrigo S, Cameron BA, Madl J, Kaltenbacher W, Kok T, Zgierski-Johnston CM, Peyronnet R, Kohl P, Sacconi L, Rog-Zielinska EA. Different effects of cardiomyocyte contractile activity on transverse and axial tubular system luminal content dynamics. J Mol Cell Cardiol 2024:S0022-2828(24)00181-0. [PMID: 39491670 DOI: 10.1016/j.yjmcc.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/06/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Efficient excitation-contraction coupling of mammalian ventricular cardiomyocytes depends on the transverse - axial tubular system (TATS), a network of surface membrane invaginations. TATS enables tight coupling of sarcolemmal and sarcoplasmic reticulum membranes, which is essential for rapid Ca2+-induced Ca2+ release, and uniform contraction upon electrical stimulation. The majority of TATS in healthy ventricular cardiomyocytes is composed of transverse tubules (TT, ~90 % of TATS in rabbit). The remainder consists of mostly axial tubules (AT), which are less abundant and less well studied. In disease, however, the relative abundance of TT and AT changes. The mechanisms and relevance of this change are not known, and understanding them requires a more targeted effort to study the dynamics of AT structure and function. While TATS content is continuous with the interstitial space, it is contained within a domain of restricted diffusion. We have previously shown that TT are cyclically squeezed during stretch and contraction. This can contribute to TT content mixing and accelerates luminal content exchange with the environment. Here, we explore the effects of cardiomyocyte stretch and contraction on AT. METHODS TATS structure and diffusion dynamics were studied using 3D electron tomography of rabbit left ventricular cardiomyocytes, preserved at rest or during contraction, and ventricular tissue preserved at rest or during stretch, as well as live-cell TATS content exchange measurements. RESULTS We show (i) that cardiomyocyte contraction is associated with an increase in the apparent speed of diffusion of TT content that scales with beating rate and degree of cell shortening. In contrast, (ii) AT develop membrane folds and constrictions during contraction, (iii) with no effect of contraction on luminal exchange dynamics, while (iv) cardiomyocyte stretch is associated with AT straightening and AT and TT 'squeezing' that (v) supports an acceleration of the apparent speed of diffusion in AT and TT. Finally, (vi) we present a simple computational model outlining the potential relevance of AT in healthy and diseased cells. CONCLUSIONS Our results indicate that TT and AT are differently affected by the cardiac contractile cycle, and suggest that AT may play a role in ensuring TATS network content homogeneity in diseased cardiomyocytes. Further research is needed to explore the interplay of structural and functional remodelling of different TATS components in failing myocardium.
Collapse
Affiliation(s)
- J Greiner
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany; Centre for Integrative Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - M Dente
- Department of Experimental and Clinical Medicine, Division of Physiology, University of Florence, Florence, Italy
| | - S Orós-Rodrigo
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - B A Cameron
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Madl
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - W Kaltenbacher
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - T Kok
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - C M Zgierski-Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - R Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - P Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany; Centre for Integrative Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany; Faculty of Engineering, University of Freiburg, Freiburg, Germany
| | - L Sacconi
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Clinical Physiology, National Research Council, Florence, Italy
| | - E A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany; Centre for Integrative Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
2
|
Lahiri SK, Lu J, Aguilar-Sanchez Y, Li H, Moreira LM, Hulsurkar MM, Mendoza A, Turkieltaub Paredes MR, Navarro-Garcia JA, Munivez E, Horist B, Moore OM, Weninger G, Brandenburg S, Lenz C, Lehnart SE, Sayeed R, Krasopoulos G, Srivastava V, Zhang L, Karch JM, Reilly S, Wehrens XHT. Targeting calpain-2-mediated junctophilin-2 cleavage delays heart failure progression following myocardial infarction. J Mol Cell Cardiol 2024; 194:85-95. [PMID: 38960317 PMCID: PMC11519832 DOI: 10.1016/j.yjmcc.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 06/18/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
Coronary heart disease (CHD) is a prevalent cardiac disease that causes over 370,000 deaths annually in the USA. In CHD, occlusion of a coronary artery causes ischemia of the cardiac muscle, which results in myocardial infarction (MI). Junctophilin-2 (JPH2) is a membrane protein that ensures efficient calcium handling and proper excitation-contraction coupling. Studies have identified loss of JPH2 due to calpain-mediated proteolysis as a key pathogenic event in ischemia-induced heart failure (HF). Our findings show that calpain-2-mediated JPH2 cleavage yields increased levels of a C-terminal cleaved peptide (JPH2-CTP) in patients with ischemic cardiomyopathy and mice with experimental MI. We created a novel knock-in mouse model by removing residues 479-SPAGTPPQ-486 to prevent calpain-2-mediated cleavage at this site. Functional and molecular assessment of cardiac function post-MI in cleavage site deletion (CSD) mice showed preserved cardiac contractility and reduced dilation, reduced JPH2-CTP levels, attenuated adverse remodeling, improved T-tubular structure, and normalized SR Ca2+-handling. Adenovirus mediated calpain-2 knockdown in mice exhibited similar findings. Pulldown of CTP followed by proteomic analysis revealed valosin-containing protein (VCP) and BAG family molecular chaperone regulator 3 (BAG3) as novel binding partners of JPH2. Together, our findings suggest that blocking calpain-2-mediated JPH2 cleavage may be a promising new strategy for delaying the development of HF following MI.
Collapse
Affiliation(s)
- Satadru K Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Jiao Lu
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Hui Li
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lucia M Moreira
- Cardiovascular Medicine, Radcliffe Dept of Medicine, University of Oxford, UK
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Arielys Mendoza
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Mara R Turkieltaub Paredes
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Jose Alberto Navarro-Garcia
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Brooke Horist
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Oliver M Moore
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Sören Brandenburg
- Department of Cardiology & Pneumology, Heart Research Center Göttingen; Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Christof Lenz
- Department of Clinical Chemistry, University Medical Center Göttingen, Germany; Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stephan E Lehnart
- Department of Cardiology & Pneumology, Heart Research Center Göttingen; Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Rana Sayeed
- Cardiothoracic Unit, John Radcliffe Hospital, Oxford, UK
| | | | | | - Lilei Zhang
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jason M Karch
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Svetlana Reilly
- Cardiovascular Medicine, Radcliffe Dept of Medicine, University of Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine/Cardiology, Baylor College of Medicine, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Paulke NJ, Fleischhacker C, Wegener JB, Riedemann GC, Cretu C, Mushtaq M, Zaremba N, Möbius W, Zühlke Y, Wedemeyer J, Liebmann L, Gorshkova AA, Kownatzki-Danger D, Wagner E, Kohl T, Wichmann C, Jahn O, Urlaub H, Toischer K, Hasenfuß G, Moser T, Preobraschenski J, Lenz C, Rog-Zielinska EA, Lehnart SE, Brandenburg S. Dysferlin Enables Tubular Membrane Proliferation in Cardiac Hypertrophy. Circ Res 2024; 135:554-574. [PMID: 39011635 DOI: 10.1161/circresaha.124.324588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Cardiac hypertrophy compensates for increased biomechanical stress of the heart induced by prevalent cardiovascular pathologies but can result in heart failure if left untreated. Here, we hypothesized that the membrane fusion and repair protein dysferlin is critical for the integrity of the transverse-axial tubule (TAT) network inside cardiomyocytes and contributes to the proliferation of TAT endomembranes during pressure overload-induced cardiac hypertrophy. METHODS Stimulated emission depletion and electron microscopy were used to localize dysferlin in mouse and human cardiomyocytes. Data-independent acquisition mass spectrometry revealed the cardiac dysferlin interactome and proteomic changes of the heart in dysferlin-knockout mice. After transverse aortic constriction, we compared the hypertrophic response of wild-type versus dysferlin-knockout hearts and studied TAT network remodeling mechanisms inside cardiomyocytes by live-cell membrane imaging. RESULTS We localized dysferlin in a vesicular compartment in nanometric proximity to contact sites of the TAT network with the sarcoplasmic reticulum, a.k.a. junctional complexes for Ca2+-induced Ca2+ release. Interactome analyses demonstrated a novel protein interaction of dysferlin with the membrane-tethering sarcoplasmic reticulum protein juncophilin-2, a putative interactor of L-type Ca2+ channels and ryanodine receptor Ca2+ release channels in junctional complexes. Although the dysferlin-knockout caused a mild progressive phenotype of dilated cardiomyopathy, global proteome analysis revealed changes preceding systolic failure. Following transverse aortic constriction, dysferlin protein expression was significantly increased in hypertrophied wild-type myocardium, while dysferlin-knockout animals presented markedly reduced left-ventricular hypertrophy. Live-cell membrane imaging showed a profound reorganization of the TAT network in wild-type left-ventricular myocytes after transverse aortic constriction with robust proliferation of axial tubules, which critically depended on the increased expression of dysferlin within newly emerging tubule components. CONCLUSIONS Dysferlin represents a new molecular target in cardiac disease that protects the integrity of tubule-sarcoplasmic reticulum junctional complexes for regulated excitation-contraction coupling and controls TAT network reorganization and tubular membrane proliferation in cardiomyocyte hypertrophy induced by pressure overload.
Collapse
Affiliation(s)
- Nora Josefine Paulke
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Carolin Fleischhacker
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Justus B Wegener
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Gabriel C Riedemann
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Constantin Cretu
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience and InnerEarLab (C.C., J.P.), University Medical Center Göttingen, Germany
| | - Mufassra Mushtaq
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Nina Zaremba
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Electron Microscopy, City Campus (W.M.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Yannik Zühlke
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Jasper Wedemeyer
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Lorenz Liebmann
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Anastasiia A Gorshkova
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Daniel Kownatzki-Danger
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Now with Institute of Transfusion Medicine, University Hospital Schleswig-Holstein; Kiel, Germany (D.K.-D)
| | - Eva Wagner
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Tobias Kohl
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab and Center for Biostructural Imaging of Neurodegeneration (C.W.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Olaf Jahn
- Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy (O.J.), University Medical Center Göttingen, Germany
- Neuroproteomics Group, Department of Molecular Neurobiology (O.J.)
| | - Henning Urlaub
- Department of Clinical Chemistry (H.U., C.L.), University Medical Center Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany (H.U., C.L.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Karl Toischer
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany (K.T., G.H., S.E.L.)
| | - Gerd Hasenfuß
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany (K.T., G.H., S.E.L.)
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab (T.M.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Julia Preobraschenski
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience and InnerEarLab (C.C., J.P.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Christof Lenz
- Department of Clinical Chemistry (H.U., C.L.), University Medical Center Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany (H.U., C.L.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Germany (E.A.R.-Z.)
| | - Stephan E Lehnart
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany (K.T., G.H., S.E.L.)
| | - Sören Brandenburg
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| |
Collapse
|
4
|
Hord JM, Anderson ME, Prouty SJ, Melton S, Gastel Z, Zimmerman K, Weiss RM, Campbell KP. Matriglycan maintains t-tubule structural integrity in cardiac muscle. Proc Natl Acad Sci U S A 2024; 121:e2402890121. [PMID: 38771868 PMCID: PMC11145246 DOI: 10.1073/pnas.2402890121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/08/2024] [Indexed: 05/23/2024] Open
Abstract
Maintaining the structure of cardiac membranes and membrane organelles is essential for heart function. A critical cardiac membrane organelle is the transverse tubule system (called the t-tubule system) which is an invagination of the surface membrane. A unique structural characteristic of the cardiac muscle t-tubule system is the extension of the extracellular matrix (ECM) from the surface membrane into the t-tubule lumen. However, the importance of the ECM extending into the cardiac t-tubule lumen is not well understood. Dystroglycan (DG) is an ECM receptor in the surface membrane of many cells, and it is also expressed in t-tubules in cardiac muscle. Extensive posttranslational processing and O-glycosylation are required for DG to bind ECM proteins and the binding is mediated by a glycan structure known as matriglycan. Genetic disruption resulting in defective O-glycosylation of DG results in muscular dystrophy with cardiorespiratory pathophysiology. Here, we show that DG is essential for maintaining cardiac t-tubule structural integrity. Mice with defects in O-glycosylation of DG developed normal t-tubules but were susceptible to stress-induced t-tubule loss or severing that contributed to cardiac dysfunction and disease progression. Finally, we observed similar stress-induced cardiac t-tubule disruption in a cohort of mice that solely lacked matriglycan. Collectively, our data indicate that DG in t-tubules anchors the luminal ECM to the t-tubule membrane via the polysaccharide matriglycan, which is critical to transmitting structural strength of the ECM to the t-tubules and provides resistance to mechanical stress, ultimately preventing disruptions in cardiac t-tubule integrity.
Collapse
Affiliation(s)
- Jeffrey M. Hord
- HHMI, University of Iowa, Iowa City, IA52242
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Mary E. Anderson
- HHMI, University of Iowa, Iowa City, IA52242
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Sally J. Prouty
- HHMI, University of Iowa, Iowa City, IA52242
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Shelly Melton
- HHMI, University of Iowa, Iowa City, IA52242
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Zeita Gastel
- HHMI, University of Iowa, Iowa City, IA52242
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Kathy Zimmerman
- Division of Cardiology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Robert M. Weiss
- Division of Cardiology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Abboud Cardiovascular Research Center, Carver College of Medicine, Department of Internal Medicine-Cardiovascular Medicine, University of Iowa, Iowa City, IA52242
- Iowa City Veterans Affairs Health Care System, University of Iowa, Iowa City, IA52242
| | - Kevin P. Campbell
- HHMI, University of Iowa, Iowa City, IA52242
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| |
Collapse
|
5
|
Moammer H, Bai J, Jones TLM, Ward M, Barrett C, Crossman DJ. Pirfenidone increases transverse tubule length in the infarcted rat myocardium. Interface Focus 2023; 13:20230047. [PMID: 38106917 PMCID: PMC10722216 DOI: 10.1098/rsfs.2023.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
Transverse (t)-tubule remodelling is a prominent feature of heart failure with reduced ejection fraction (HFrEF). In our previous research, we identified an increased amount of collagen within the t-tubules of HFrEF patients, suggesting fibrosis could contribute to the remodelling of t-tubules. In this research, we tested this hypothesis in a rodent model of myocardial infarction induced heart failure that was treated with the anti-fibrotic pirfenidone. Confocal microscopy demonstrated loss of t-tubules within the border zone region of the infarct. This was documented as a reduction in t-tubule frequency, area, length, and transverse elements. Eight weeks of pirfenidone treatment was able to significantly increase the area and length of the t-tubules within the border zone. Echocardiography showed no improvement with pirfenidone treatment. Surprisingly, pirfenidone significantly increased the thickness of the t-tubules in the remote left ventricle of heart failure animals. Dilation of t-tubules is a common feature in heart failure suggesting this may negatively impact function but there was no functional loss associated with pirfenidone treatment. However, due to the relatively short duration of treatment compared to that used clinically, the impact of long-term treatment on t-tubule structure should be investigated in future studies.
Collapse
Affiliation(s)
- Hussam Moammer
- Manaaki Manawa—The Centre for Heart Research, Department of Physiology, School of Medical and Health Sciences, Faculty of Medical and Health Sciences, Waipapa Taumata Rau / The University of Auckland, Park Road, Grafton, Auckland, New Zealand
- Department of Clinical Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jizhong Bai
- Manaaki Manawa—The Centre for Heart Research, Department of Physiology, School of Medical and Health Sciences, Faculty of Medical and Health Sciences, Waipapa Taumata Rau / The University of Auckland, Park Road, Grafton, Auckland, New Zealand
| | - Timothy L. M. Jones
- Manaaki Manawa—The Centre for Heart Research, Department of Physiology, School of Medical and Health Sciences, Faculty of Medical and Health Sciences, Waipapa Taumata Rau / The University of Auckland, Park Road, Grafton, Auckland, New Zealand
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marie Ward
- Manaaki Manawa—The Centre for Heart Research, Department of Physiology, School of Medical and Health Sciences, Faculty of Medical and Health Sciences, Waipapa Taumata Rau / The University of Auckland, Park Road, Grafton, Auckland, New Zealand
| | - Caroyln Barrett
- Manaaki Manawa—The Centre for Heart Research, Department of Physiology, School of Medical and Health Sciences, Faculty of Medical and Health Sciences, Waipapa Taumata Rau / The University of Auckland, Park Road, Grafton, Auckland, New Zealand
| | - David J. Crossman
- Manaaki Manawa—The Centre for Heart Research, Department of Physiology, School of Medical and Health Sciences, Faculty of Medical and Health Sciences, Waipapa Taumata Rau / The University of Auckland, Park Road, Grafton, Auckland, New Zealand
| |
Collapse
|
6
|
Waddell HMM, Mereacre V, Alvarado FJ, Munro ML. Clustering properties of the cardiac ryanodine receptor in health and heart failure. J Mol Cell Cardiol 2023; 185:38-49. [PMID: 37890552 PMCID: PMC10717225 DOI: 10.1016/j.yjmcc.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The cardiac ryanodine receptor (RyR2) is an intracellular Ca2+ release channel vital for the function of the heart. Physiologically, RyR2 is triggered to release Ca2+ from the sarcoplasmic reticulum (SR) which enables cardiac contraction; however, spontaneous Ca2+ leak from RyR2 has been implicated in the pathophysiology of heart failure (HF). RyR2 channels have been well documented to assemble into clusters within the SR membrane, with the organisation of RyR2 clusters recently gaining interest as a mechanism by which the occurrence of pathological Ca2+ leak is regulated, including in HF. In this review, we explain the terminology relating to key nanoscale RyR2 clustering properties as both single clusters and functionally grouped Ca2+ release units, with a focus on the advancements in super-resolution imaging approaches which have enabled the detailed study of cluster organisation. Further, we discuss proposed mechanisms for modulating RyR2 channel organisation and the debate regarding the potential impact of cluster organisation on Ca2+ leak activity. Finally, recent experimental evidence investigating the nanoscale remodelling and functional alterations of RyR2 clusters in HF is discussed with consideration of the clinical implications.
Collapse
Affiliation(s)
- Helen M M Waddell
- Department of Physiology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Valeria Mereacre
- Department of Physiology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Francisco J Alvarado
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Michelle L Munro
- Department of Physiology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
7
|
Dries E, Gilbert G, Roderick HL, Sipido KR. The ryanodine receptor microdomain in cardiomyocytes. Cell Calcium 2023; 114:102769. [PMID: 37390591 DOI: 10.1016/j.ceca.2023.102769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
The ryanodine receptor type 2 (RyR) is a key player in Ca2+ handling during excitation-contraction coupling. During each heartbeat, RyR channels are responsible for linking the action potential with the contractile machinery of the cardiomyocyte by releasing Ca2+ from the sarcoplasmic reticulum. RyR function is fine-tuned by associated signalling molecules, arrangement in clusters and subcellular localization. These parameters together define RyR function within microdomains and are subject to disease remodelling. This review describes the latest findings on RyR microdomain organization, the alterations with disease which result in increased subcellular heterogeneity and emergence of microdomains with enhanced arrhythmogenic potential, and presents novel technologies that guide future research to study and target RyR channels within specific microdomains.
Collapse
Affiliation(s)
- Eef Dries
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Guillaume Gilbert
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Laboratoire ORPHY EA 4324, Université de Brest, Brest, France
| | - H Llewelyn Roderick
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Karin R Sipido
- Lab of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Alvarez JAE, Jafri MS, Ullah A. Local Control Model of a Human Ventricular Myocyte: An Exploration of Frequency-Dependent Changes and Calcium Sparks. Biomolecules 2023; 13:1259. [PMID: 37627324 PMCID: PMC10452762 DOI: 10.3390/biom13081259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Calcium (Ca2+) sparks are the elementary events of excitation-contraction coupling, yet they are not explicitly represented in human ventricular myocyte models. A stochastic ventricular cardiomyocyte human model that adapts to intracellular Ca2+ ([Ca2+]i) dynamics, spark regulation, and frequency-dependent changes in the form of locally controlled Ca2+ release was developed. The 20,000 CRUs in this model are composed of 9 individual LCCs and 49 RyRs that function as couplons. The simulated action potential duration at 1 Hz steady-state pacing is ~0.280 s similar to human ventricular cell recordings. Rate-dependence experiments reveal that APD shortening mechanisms are largely contributed by the L-type calcium channel inactivation, RyR open fraction, and [Ca2+]myo concentrations. The dynamic slow-rapid-slow pacing protocol shows that RyR open probability during high pacing frequency (2.5 Hz) switches to an adapted "nonconducting" form of Ca2+-dependent transition state. The predicted force was also observed to be increased in high pacing, but the SR Ca2+ fractional release was lower due to the smaller difference between diastolic and systolic [Ca2+]SR. Restitution analysis through the S1S2 protocol and increased LCC Ca2+-dependent activation rate show that the duration of LCC opening helps modulate its effects on the APD restitution at different diastolic intervals. Ultimately, a longer duration of calcium sparks was observed in relation to the SR Ca2+ loading at high pacing rates. Overall, this study demonstrates the spontaneous Ca2+ release events and ion channel responses throughout various stimuli.
Collapse
Affiliation(s)
| | - M. Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
| |
Collapse
|
9
|
White D, Smith MA, Chanda B, Goldsmith RH. Strategies for Overcoming the Single-Molecule Concentration Barrier. ACS MEASUREMENT SCIENCE AU 2023; 3:239-257. [PMID: 37600457 PMCID: PMC10436376 DOI: 10.1021/acsmeasuresciau.3c00002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 08/22/2023]
Abstract
Fluorescence-based single-molecule approaches have helped revolutionize our understanding of chemical and biological mechanisms. Unfortunately, these methods are only suitable at low concentrations of fluorescent molecules so that single fluorescent species of interest can be successfully resolved beyond background signal. The application of these techniques has therefore been limited to high-affinity interactions despite most biological and chemical processes occurring at much higher reactant concentrations. Fortunately, recent methodological advances have demonstrated that this concentration barrier can indeed be broken, with techniques reaching concentrations as high as 1 mM. The goal of this Review is to discuss the challenges in performing single-molecule fluorescence techniques at high-concentration, offer applications in both biology and chemistry, and highlight the major milestones that shatter the concentration barrier. We also hope to inspire the widespread use of these techniques so we can begin exploring the new physical phenomena lying beyond this barrier.
Collapse
Affiliation(s)
- David
S. White
- Department
of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Mackinsey A. Smith
- Department
of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Baron Chanda
- Center
for
Investigation of Membrane Excitability Diseases, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Randall H. Goldsmith
- Department
of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
10
|
Tamkus G, Uchida K, Lopatin AN. T-tubule recovery after detubulation in isolated mouse cardiomyocytes. Physiol Rep 2023; 11:e15779. [PMID: 37537144 PMCID: PMC10400551 DOI: 10.14814/phy2.15779] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 08/05/2023] Open
Abstract
Remodeling of cardiac t-tubules in normal and pathophysiological conditions is an important process contributing to the functional performance of the heart. While it is well documented that deterioration of t-tubule network associated with various pathological conditions can be reversed under certain conditions, the mechanistic understanding of the recovery process is essentially lacking. Accordingly, in this study we investigated some aspects of the recovery of t-tubules after experimentally-induced detubulation. T-tubules of isolated mouse ventricular myocytes were first sealed using osmotic shock approach, and their recovery under various experimental conditions was then characterized using electrophysiologic and imaging techniques. The data show that t-tubule recovery is a strongly temperature-dependent process involving reopening of previously collapsed t-tubular segments. T-tubule recovery is slowed by (1) metabolic inhibition of cells, (2) reducing influx of extracellular Ca2+ as well as by (3) both stabilization and disruption of microtubules. Overall, the data show that t-tubule recovery is a highly dynamic process involving several central intracellular structures and processes and lay the basis for more detailed investigations in this area.
Collapse
Affiliation(s)
- Greta Tamkus
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Present address:
John T. Milliken Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Keita Uchida
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Present address:
Department of PhysiologyPennsylvania Muscle Institute, University of Pennsylvania, Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Anatoli N. Lopatin
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
11
|
Xu X, Elkenani M, Tan X, Hain JK, Cui B, Schnelle M, Hasenfuss G, Toischer K, Mohamed BA. DNA Methylation Analysis Identifies Novel Epigenetic Loci in Dilated Murine Heart upon Exposure to Volume Overload. Int J Mol Sci 2023; 24:ijms24065885. [PMID: 36982963 PMCID: PMC10059258 DOI: 10.3390/ijms24065885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Left ventricular (LV) dilatation, a prominent risk factor for heart failure (HF), precedes functional deterioration and is used to stratify patients at risk for arrhythmias and cardiac mortality. Aberrant DNA methylation contributes to maladaptive cardiac remodeling and HF progression following pressure overload and ischemic cardiac insults. However, no study has examined cardiac DNA methylation upon exposure to volume overload (VO) despite being relatively common among HF patients. We carried out global methylome analysis of LV harvested at a decompensated HF stage following exposure to VO induced by aortocaval shunt. VO resulted in pathological cardiac remodeling, characterized by massive LV dilatation and contractile dysfunction at 16 weeks after shunt. Although methylated DNA was not markedly altered globally, 25 differentially methylated promoter regions (DMRs) were identified in shunt vs. sham hearts (20 hypermethylated and 5 hypomethylated regions). The validated hypermethylated loci in Junctophilin-2 (Jph2), Signal peptidase complex subunit 3 (Spcs3), Vesicle-associated membrane protein-associated protein B (Vapb), and Inositol polyphosphate multikinase (Ipmk) were associated with the respective downregulated expression and were consistently observed in dilated LV early after shunt at 1 week after shunt, before functional deterioration starts to manifest. These hypermethylated loci were also detected peripherally in the blood of the shunt mice. Altogether, we have identified conserved DMRs that could be novel epigenetic biomarkers in dilated LV upon VO exposure.
Collapse
Affiliation(s)
- Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
| | - Manar Elkenani
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Xiaoying Tan
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
- Department of Nephrology and Rheumatology, University Medical Center of Göttingen, 37075 Göttingen, Germany
| | - Jara Katharina Hain
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Baolong Cui
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
| | - Moritz Schnelle
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), 37075 Göttingen, Germany
| |
Collapse
|
12
|
McCoy MD, Ullah A, Lederer WJ, Jafri MS. Understanding Calmodulin Variants Affecting Calcium-Dependent Inactivation of L-Type Calcium Channels through Whole-Cell Simulation of the Cardiac Ventricular Myocyte. Biomolecules 2022; 13:72. [PMID: 36671457 PMCID: PMC9855640 DOI: 10.3390/biom13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Mutations in the calcium-sensing protein calmodulin (CaM) have been linked to two cardiac arrhythmia diseases, Long QT Syndrome 14 (LQT14) and Catecholaminergic Polymorphic Ventricular Tachycardia Type 4 (CPVT4), with varying degrees of severity. Functional characterization of the CaM mutants most strongly associated with LQT14 show a clear disruption of the calcium-dependent inactivation (CDI) of the L-Type calcium channel (LCC). CPVT4 mutants on the other hand are associated with changes in their affinity to the ryanodine receptor. In clinical studies, some variants have been associated with both CPVT4 and LQT15. This study uses simulations in a model for excitation-contraction coupling in the rat ventricular myocytes to understand how LQT14 variant might give the functional phenotype similar to CPVT4. Changing the CaM-dependent transition rate by a factor of 0.75 corresponding to the D96V variant and by a factor of 0.90 corresponding to the F142L or N98S variants, in a physiologically based stochastic model of the LCC prolonger, the action potential duration changed by a small amount in a cardiac myocyte but did not disrupt CICR at 1, 2, and 4 Hz. Under beta-adrenergic simulation abnormal excitation-contraction coupling was observed above 2 Hz pacing for the mutant CaM. The same conditions applied under beta-adrenergic stimulation led to the rapid onset of arrhythmia in the mutant CaM simulations. Simulations with the LQT14 mutations under the conditions of rapid pacing with beta-adrenergic stimulation drives the cardiac myocyte toward an arrhythmic state known as Ca2+ overload. These simulations provide a mechanistic link to a disease state for LQT14-associated mutations in CaM to yield a CPVT4 phenotype. The results show that small changes to the CaM-regulated inactivation of LCC promote arrhythmia and underscore the significance of CDI in proper heart function.
Collapse
Affiliation(s)
- Matthew D. McCoy
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Innovation Center for Biomedical Informatics, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| | - W. Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - M. Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| |
Collapse
|
13
|
Paudel R, Jafri MS, Ullah A. Pacing Dynamics Determines the Arrhythmogenic Mechanism of the CPVT2-Causing CASQ2 G112+5X Mutation in a Guinea Pig Ventricular Myocyte Computational Model. Genes (Basel) 2022; 14:23. [PMID: 36672764 PMCID: PMC9858930 DOI: 10.3390/genes14010023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Calsequestrin Type 2 (CASQ2) is a high-capacity, low-affinity, Ca2+-binding protein expressed in the sarcoplasmic reticulum (SR) of the cardiac myocyte. Mutations in CASQ2 have been linked to the arrhythmia catecholaminergic polymorphic ventricular tachycardia (CPVT2) that occurs with acute emotional stress or exercise can result in sudden cardiac death (SCD). CASQ2G112+5X is a 16 bp (339-354) deletion CASQ2 mutation that prevents the protein expression due to premature stop codon. Understanding the subcellular mechanisms of CPVT2 is experimentally challenging because the occurrence of arrhythmia is rare. To obtain an insight into the characteristics of this rare disease, simulation studies using a local control stochastic computational model of the Guinea pig ventricular myocyte investigated how the mutant CASQ2s may be responsible for the development of an arrhythmogenic episode under the condition of β-adrenergic stimulation or in the slowing of heart rate afterward once β-adrenergic stimulation ceases. Adjustment of the computational model parameters based upon recent experiments explore the functional changes caused by the CASQ2 mutation. In the simulation studies under rapid pacing (6 Hz), electromechanically concordant cellular alternans appeared under β-adrenergic stimulation in the CPVT mutant but not in the wild-type nor in the non-β-stimulated mutant. Similarly, the simulations of accelerating pacing from slow to rapid and back to the slow pacing did not display alternans but did generate early afterdepolarizations (EADs) during the period of second slow pacing subsequent acceleration of rapid pacing.
Collapse
Affiliation(s)
- Roshan Paudel
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
14
|
Brandenburg S, Drews L, Schönberger HL, Jacob CF, Paulke NJ, Beuthner BE, Topci R, Kohl T, Neuenroth L, Kutschka I, Urlaub H, Kück F, Leha A, Friede T, Seidler T, Jacobshagen C, Toischer K, Puls M, Hasenfuß G, Lenz C, Lehnart SE. Direct proteomic and high-resolution microscopy biopsy analysis identifies distinct ventricular fates in severe aortic stenosis. J Mol Cell Cardiol 2022; 173:1-15. [PMID: 36084744 DOI: 10.1016/j.yjmcc.2022.08.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/03/2022] [Accepted: 08/31/2022] [Indexed: 01/06/2023]
Abstract
The incidence of aortic valve stenosis (AS), the most common reason for aortic valve replacement (AVR), increases with population ageing. While untreated AS is associated with high mortality, different hemodynamic subtypes range from normal left-ventricular function to severe heart failure. However, the molecular nature underlying four different AS subclasses, suggesting vastly different myocardial fates, is unknown. Here, we used direct proteomic analysis of small left-ventricular biopsies to identify unique protein expression profiles and subtype-specific AS mechanisms. Left-ventricular endomyocardial biopsies were harvested from patients during transcatheter AVR, and inclusion criteria were based on echocardiographic diagnosis of severe AS and guideline-defined AS-subtype classification: 1) normal ejection fraction (EF)/high-gradient; 2) low EF/high-gradient; 3) low EF/low-gradient; and 4) paradoxical low-flow/low-gradient AS. Samples from non-failing donor hearts served as control. We analyzed 25 individual left-ventricular biopsies by data-independent acquisition mass spectrometry (DIA-MS), and 26 biopsies by histomorphology and cardiomyocytes by STimulated Emission Depletion (STED) superresolution microscopy. Notably, DIA-MS reliably detected 2273 proteins throughout each individual left-ventricular biopsy, of which 160 proteins showed significant abundance changes between AS-subtype and non-failing samples including the cardiac ryanodine receptor (RyR2). Hierarchical clustering segregated unique proteotypes that identified three hemodynamic AS-subtypes. Additionally, distinct proteotypes were linked with AS-subtype specific differences in cardiomyocyte hypertrophy. Furthermore, superresolution microscopy of immunolabeled biopsy sections showed subcellular RyR2-cluster fragmentation and disruption of the functionally important association with transverse tubules, which occurred specifically in patients with systolic dysfunction and may hence contribute to depressed left-ventricular function in AS.
Collapse
Affiliation(s)
- Sören Brandenburg
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany; Collaborative Research Center SFB1002 "Modulatory Units in Heart Failure", University of Göttingen, Germany.
| | - Lena Drews
- Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Hanne-Lea Schönberger
- Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Christoph F Jacob
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Nora Josefine Paulke
- Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Bo E Beuthner
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Rodi Topci
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany
| | - Tobias Kohl
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Lisa Neuenroth
- Department of Clinical Chemistry, University Medical Center Göttingen, Germany
| | - Ingo Kutschka
- Clinic of Cardiothoracic & Vascular Surgery, University Medical Center Göttingen, Germany
| | - Henning Urlaub
- Department of Clinical Chemistry, University Medical Center Göttingen, Germany; Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, Germany
| | - Fabian Kück
- Department of Medical Statistics, University Medical Center Göttingen, Germany
| | - Andreas Leha
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany; Department of Medical Statistics, University Medical Center Göttingen, Germany
| | - Tim Friede
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany; Department of Medical Statistics, University Medical Center Göttingen, Germany
| | - Tim Seidler
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Claudius Jacobshagen
- Department of Cardiology, Intensive Care & Angiology, Vincentius-Diakonissen-Hospital Karlsruhe, Germany
| | - Karl Toischer
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany; Collaborative Research Center SFB1002 "Modulatory Units in Heart Failure", University of Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| | - Miriam Puls
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Gerd Hasenfuß
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany; Collaborative Research Center SFB1002 "Modulatory Units in Heart Failure", University of Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| | - Christof Lenz
- Collaborative Research Center SFB1002 "Modulatory Units in Heart Failure", University of Göttingen, Germany; Department of Clinical Chemistry, University Medical Center Göttingen, Germany; Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany; Leducq Transatlantic Network of Excellence CURE-PLaN, Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany
| | - Stephan E Lehnart
- Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany; Cellular Biophysics & Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany; Collaborative Research Center SFB1002 "Modulatory Units in Heart Failure", University of Göttingen, Germany; Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany; Leducq Transatlantic Network of Excellence CURE-PLaN, Clinic of Cardiology & Pneumology, University Medical Center Göttingen, Germany.
| |
Collapse
|
15
|
Perera T, Pius C, Niort B, Radcliffe EJ, Dibb KM, Trafford AW, Pinali C. Serial block face scanning electron microscopy reveals region-dependent remodelling of transverse tubules post-myocardial infarction. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210331. [PMID: 36189812 PMCID: PMC9527908 DOI: 10.1098/rstb.2021.0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 11/18/2022] Open
Abstract
The highly organized transverse tubule (t-tubule) network facilitates cardiac excitation-contraction coupling and synchronous cardiac myocyte contraction. In cardiac failure secondary to myocardial infarction (MI), changes in the structure and organization of t-tubules result in impaired cardiac contractility. However, there is still little knowledge on the regional variation of t-tubule remodelling in cardiac failure post-MI. Here, we investigate post-MI t-tubule remodelling in infarct border and remote regions, using serial block face scanning electron microscopy (SBF-SEM) applied to a translationally relevant sheep ischaemia reperfusion MI model and matched controls. We performed minimally invasive coronary angioplasty of the left anterior descending artery, followed by reperfusion after 90 min to establish the MI model. Left ventricular tissues obtained from control and MI hearts eight weeks post-MI were imaged using SBF-SEM. Image analysis generated three-dimensional reconstructions of the t-tubular network in control, MI border and remote regions. Quantitative analysis revealed that the MI border region was characterized by t-tubule depletion and fragmentation, dilation of surviving t-tubules and t-tubule elongation. This study highlights region-dependent remodelling of the tubular network post-MI and may provide novel localized therapeutic targets aimed at preservation or restoration of the t-tubules to manage cardiac contractility post-MI. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Tharushi Perera
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Charlene Pius
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Barbara Niort
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Emma J. Radcliffe
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Katharine M. Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Andrew W. Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| | - Christian Pinali
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, 46 Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
16
|
Paudel R, Jafri MS, Ullah A. The Role of Ca 2+ Sparks in Force Frequency Relationships in Guinea Pig Ventricular Myocytes. Biomolecules 2022; 12:1577. [PMID: 36358926 PMCID: PMC9687237 DOI: 10.3390/biom12111577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 10/13/2023] Open
Abstract
Calcium sparks are the elementary Ca2+ release events in excitation-contraction coupling that underlie the Ca2+ transient. The frequency-dependent contractile force generated by cardiac myocytes depends upon the characteristics of the Ca2+ transients. A stochastic computational local control model of a guinea pig ventricular cardiomyocyte was developed, to gain insight into mechanisms of force-frequency relationship (FFR). This required the creation of a new three-state RyR2 model that reproduced the adaptive behavior of RyR2, in which the RyR2 channels transition into a different state when exposed to prolonged elevated subspace [Ca2+]. The model simulations agree with previous experimental and modeling studies on interval-force relations. Unlike previous common pool models, this local control model displayed stable action potential trains at 7 Hz. The duration and the amplitude of the [Ca2+]myo transients increase in pacing rates consistent with the experiments. The [Ca2+]myo transient reaches its peak value at 4 Hz and decreases afterward, consistent with experimental force-frequency curves. The model predicts, in agreement with previous modeling studies of Jafri and co-workers, diastolic sarcoplasmic reticulum, [Ca2+]sr, and RyR2 adaptation increase with the increased stimulation frequency, producing rising, rather than falling, amplitude of the myoplasmic [Ca2+] transients. However, the local control model also suggests that the reduction of the L-type Ca2+ current, with an increase in pacing frequency due to Ca2+-dependent inactivation, also plays a role in the negative slope of the FFR. In the simulations, the peak Ca2+ transient in the FFR correlated with the highest numbers of SR Ca2+ sparks: the larger average amplitudes of those sparks, and the longer duration of the Ca2+ sparks.
Collapse
Affiliation(s)
- Roshan Paudel
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
17
|
A new approach to the determination of tubular membrane capacitance: passive membrane electrical properties under reduced electrical conductivity of the extracellular solution. Pflugers Arch 2022; 474:1263-1274. [PMID: 36239778 PMCID: PMC9663357 DOI: 10.1007/s00424-022-02756-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/06/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
The transverse-axial tubular system (tubular system) of cardiomyocytes plays a key role in excitation–contraction coupling. To determine the area of the tubular membrane in relation to the area of the surface membrane, indirect measurements through the determination of membrane capacitances are currently used in addition to microscopic methods. Unlike existing electrophysiological methods based on an irreversible procedure (osmotic shock), the proposed new approach uses a reversible short-term intermittent increase in the electrical resistance of the extracellular medium. The resulting increase in the lumen resistance of the tubular system makes it possible to determine separate capacitances of the tubular and surface membranes. Based on the analysis of the time course of the capacitive current, computational relations were derived to quantify the elements of the electrical equivalent circuit of the measured cardiomyocyte including both capacitances. The exposition to isotonic low-conductivity sucrose solution is reversible which is the main advantage of the proposed approach allowing repetitive measurements on the same cell under control and sucrose solutions. Experiments on rat ventricular cardiomyocytes (n = 20) resulted in the surface and tubular capacitance values implying the fraction of tubular capacitance/area of 0.327 ± 0.018. We conclude that the newly proposed method provides results comparable to the data obtained by the currently used detubulation method and, in addition, by being reversible, allows repeated evaluation of surface and tubular membrane parameters on the same cell.
Collapse
|
18
|
Guarina L, Moghbel AN, Pourhosseinzadeh MS, Cudmore RH, Sato D, Clancy CE, Santana LF. Biological noise is a key determinant of the reproducibility and adaptability of cardiac pacemaking and EC coupling. J Gen Physiol 2022; 154:e202012613. [PMID: 35482009 PMCID: PMC9059386 DOI: 10.1085/jgp.202012613] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 12/23/2022] Open
Abstract
Each heartbeat begins with the generation of an action potential in pacemaking cells in the sinoatrial node. This signal triggers contraction of cardiac muscle through a process termed excitation-contraction (EC) coupling. EC coupling is initiated in dyadic structures of cardiac myocytes, where ryanodine receptors in the junctional sarcoplasmic reticulum come into close apposition with clusters of CaV1.2 channels in invaginations of the sarcolemma. Cooperative activation of CaV1.2 channels within these clusters causes a local increase in intracellular Ca2+ that activates the juxtaposed ryanodine receptors. A salient feature of healthy cardiac function is the reliable and precise beat-to-beat pacemaking and amplitude of Ca2+ transients during EC coupling. In this review, we discuss recent discoveries suggesting that the exquisite reproducibility of this system emerges, paradoxically, from high variability at subcellular, cellular, and network levels. This variability is attributable to stochastic fluctuations in ion channel trafficking, clustering, and gating, as well as dyadic structure, which increase intracellular Ca2+ variance during EC coupling. Although the effects of these large, local fluctuations in function and organization are sometimes negligible at the macroscopic level owing to spatial-temporal summation within and across cells in the tissue, recent work suggests that the "noisiness" of these intracellular Ca2+ events may either enhance or counterintuitively reduce variability in a context-dependent manner. Indeed, these noisy events may represent distinct regulatory features in the tuning of cardiac contractility. Collectively, these observations support the importance of incorporating experimentally determined values of Ca2+ variance in all EC coupling models. The high reproducibility of cardiac contraction is a paradoxical outcome of high Ca2+ signaling variability at subcellular, cellular, and network levels caused by stochastic fluctuations in multiple processes in time and space. This underlying stochasticity, which counterintuitively manifests as reliable, consistent Ca2+ transients during EC coupling, also allows for rapid changes in cardiac rhythmicity and contractility in health and disease.
Collapse
Affiliation(s)
- Laura Guarina
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Ariana Neelufar Moghbel
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | | | - Robert H. Cudmore
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Daisuke Sato
- Department of Pharmacology, University of California Davis School of Medicine, Davis, CA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Luis Fernando Santana
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| |
Collapse
|
19
|
Weninger G, Pochechueva T, El Chami D, Luo X, Kohl T, Brandenburg S, Urlaub H, Guan K, Lenz C, Lehnart SE. Calpain cleavage of Junctophilin-2 generates a spectrum of calcium-dependent cleavage products and DNA-rich NT 1-fragment domains in cardiomyocytes. Sci Rep 2022; 12:10387. [PMID: 35725601 PMCID: PMC9209451 DOI: 10.1038/s41598-022-14320-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Calpains are calcium-activated neutral proteases involved in the regulation of key signaling pathways. Junctophilin-2 (JP2) is a Calpain-specific proteolytic target and essential structural protein inside Ca2+ release units required for excitation-contraction coupling in cardiomyocytes. While downregulation of JP2 by Calpain cleavage in heart failure has been reported, the precise molecular identity of the Calpain cleavage sites and the (patho-)physiological roles of the JP2 proteolytic products remain controversial. We systematically analyzed the JP2 cleavage fragments as function of Calpain-1 versus Calpain-2 proteolytic activities, revealing that both Calpain isoforms preferentially cleave mouse JP2 at R565, but subsequently at three additional secondary Calpain cleavage sites. Moreover, we identified the Calpain-specific primary cleavage products for the first time in human iPSC-derived cardiomyocytes. Knockout of RyR2 in hiPSC-cardiomyocytes destabilized JP2 resulting in an increase of the Calpain-specific cleavage fragments. The primary N-terminal cleavage product NT1 accumulated in the nucleus of mouse and human cardiomyocytes in a Ca2+-dependent manner, closely associated with euchromatic chromosomal regions, where NT1 is proposed to function as a cardio-protective transcriptional regulator in heart failure. Taken together, our data suggest that stabilizing NT1 by preventing secondary cleavage events by Calpain and other proteases could be an important therapeutic target for future studies.
Collapse
Affiliation(s)
- Gunnar Weninger
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075, Göttingen, Germany.,Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany.,Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Tatiana Pochechueva
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075, Göttingen, Germany.,Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany
| | - Dana El Chami
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075, Göttingen, Germany.,Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Tobias Kohl
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075, Göttingen, Germany.,Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC2067), University of Göttingen, 37073, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site, 37075, Göttingen, Germany
| | - Sören Brandenburg
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075, Göttingen, Germany.,Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC2067), University of Göttingen, 37073, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site, 37075, Göttingen, Germany
| | - Henning Urlaub
- Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany.,Proteomanalyse, Department of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Christof Lenz
- Proteomanalyse, Department of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany. .,Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany.
| | - Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Str. 42a, 37075, Göttingen, Germany. .,Department of Cardiology and Pneumology, University Medical Center Göttingen, 37075, Göttingen, Germany. .,Collaborative Research Center SFB1190 "Compartmental Gates and Contact Sites in Cells", University of Göttingen, 37073, Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC2067), University of Göttingen, 37073, Göttingen, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site, 37075, Göttingen, Germany.
| |
Collapse
|
20
|
Ullah A, Hoang-Trong MT, Lederer WJ, Winslow RL, Jafri MS. Critical Requirements for the Initiation of a Cardiac Arrhythmia in Rat Ventricle: How Many Myocytes? Cells 2022; 11:cells11121878. [PMID: 35741007 PMCID: PMC9221049 DOI: 10.3390/cells11121878] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide due in a large part to arrhythmia. In order to understand how calcium dynamics play a role in arrhythmogenesis, normal and dysfunctional Ca2+ signaling in a subcellular, cellular, and tissued level is examined using cardiac ventricular myocytes at a high temporal and spatial resolution using multiscale computational modeling. Ca2+ sparks underlie normal excitation-contraction coupling. However, under pathological conditions, Ca2+ sparks can combine to form Ca2+ waves. These propagating elevations of (Ca2+)i can activate an inward Na+-Ca2+ exchanger current (INCX) that contributes to early after-depolarization (EADs) and delayed after-depolarizations (DADs). However, how cellular currents lead to full depolarization of the myocardium and how they initiate extra systoles is still not fully understood. This study explores how many myocytes must be entrained to initiate arrhythmogenic depolarizations in biophysically detailed computational models. The model presented here suggests that only a small number of myocytes must activate in order to trigger an arrhythmogenic propagating action potential. These conditions were examined in 1-D, 2-D, and 3-D considering heart geometry. The depolarization of only a few hundred ventricular myocytes is required to trigger an ectopic depolarization. The number decreases under disease conditions such as heart failure. Furthermore, in geometrically restricted parts of the heart such as the thin muscle strands found in the trabeculae and papillary muscle, the number of cells needed to trigger a propagating depolarization falls even further to less than ten myocytes.
Collapse
Affiliation(s)
- Aman Ullah
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (A.U.); (M.T.H.-T.)
| | - Minh Tuan Hoang-Trong
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (A.U.); (M.T.H.-T.)
| | - William Jonathan Lederer
- Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Raimond L. Winslow
- Institute for Computational Medicine and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 20218, USA;
- The Roux Institute, Northeastern University, Portland, ME 04102, USA
| | - Mohsin Saleet Jafri
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (A.U.); (M.T.H.-T.)
- Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Institute for Computational Medicine and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 20218, USA;
- Correspondence: ; Tel.: +1-703-993-8420
| |
Collapse
|
21
|
Švecová O, Bébarová M, Šimurdová M, Šimurda J. Fraction of the T-Tubular Membrane as an Important Parameter in Cardiac Cellular Electrophysiology: A New Way of Estimation. Front Physiol 2022; 13:837239. [PMID: 35620609 PMCID: PMC9127156 DOI: 10.3389/fphys.2022.837239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/15/2022] [Indexed: 12/02/2022] Open
Abstract
The transverse-axial tubular system (t-tubules) plays an essential role in excitation-contraction coupling in cardiomyocytes. Its remodelling is associated with various cardiac diseases. Numerous attempts were made to analyse characteristics essential for proper understanding of the t-tubules and their impact on cardiac cell function in health and disease. The currently available methodical approaches related to the fraction of the t-tubular membrane area produce diverse data. The widely used detubulation techniques cause irreversible cell impairment, thus, distinct cell samples have to be used for estimation of t-tubular parameters in untreated and detubulated cells. Our proposed alternative method is reversible and allows repetitive estimation of the fraction of t-tubular membrane (f t) in cardiomyocytes using short-term perfusion of the measured cell with a low-conductive isotonic sucrose solution. It results in a substantial increase in the electrical resistance of t-tubular lumen, thus, electrically separating the surface and t-tubular membranes. Using the whole-cell patch-clamp measurement and the new approach in enzymatically isolated rat atrial and ventricular myocytes, a set of data was measured and evaluated. The analysis of the electrical equivalent circuit resulted in the establishment of criteria for excluding measurements in which perfusion with a low conductivity solution did not affect the entire cell surface. As expected, the final average f t in ventricular myocytes (0.337 ± 0.017) was significantly higher than that in atrial myocytes (0.144 ± 0.015). The parameter f t could be estimated repetitively in a particular cell (0.345 ± 0.021 and 0.347 ± 0.023 in ventricular myocytes during the first and second sucrose perfusion, respectively). The new method is fast, simple, and leaves the measured cell intact. It can be applied in the course of experiments for which it is useful to estimate both the surface and t-tubular capacitance/area in a particular cell.
Collapse
Affiliation(s)
- Olga Švecová
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Markéta Bébarová
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Milena Šimurdová
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jiří Šimurda
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
22
|
Colman MA, Alvarez-Lacalle E, Echebarria B, Sato D, Sutanto H, Heijman J. Multi-Scale Computational Modeling of Spatial Calcium Handling From Nanodomain to Whole-Heart: Overview and Perspectives. Front Physiol 2022; 13:836622. [PMID: 35370783 PMCID: PMC8964409 DOI: 10.3389/fphys.2022.836622] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of intracellular calcium is a critical component of cardiac electrophysiology and excitation-contraction coupling. The calcium spark, the fundamental element of the intracellular calcium transient, is initiated in specialized nanodomains which co-locate the ryanodine receptors and L-type calcium channels. However, calcium homeostasis is ultimately regulated at the cellular scale, by the interaction of spatially separated but diffusively coupled nanodomains with other sub-cellular and surface-membrane calcium transport channels with strong non-linear interactions; and cardiac electrophysiology and arrhythmia mechanisms are ultimately tissue-scale phenomena, regulated by the interaction of a heterogeneous population of coupled myocytes. Recent advances in imaging modalities and image-analysis are enabling the super-resolution reconstruction of the structures responsible for regulating calcium homeostasis, including the internal structure of nanodomains themselves. Extrapolating functional and imaging data from the nanodomain to the whole-heart is non-trivial, yet essential for translational insight into disease mechanisms. Computational modeling has important roles to play in relating structural and functional data at the sub-cellular scale and translating data across the scales. This review covers recent methodological advances that enable image-based modeling of the single nanodomain and whole cardiomyocyte, as well as the development of multi-scale simulation approaches to integrate data from nanometer to whole-heart. Firstly, methods to overcome the computational challenges of simulating spatial calcium dynamics in the nanodomain are discussed, including image-based modeling at this scale. Then, recent whole-cell models, capable of capturing a range of different structures (such as the T-system and mitochondria) and cellular heterogeneity/variability are discussed at two different levels of discretization. Novel methods to integrate the models and data across the scales and simulate stochastic dynamics in tissue-scale models are then discussed, enabling elucidation of the mechanisms by which nanodomain remodeling underlies arrhythmia and contractile dysfunction. Perspectives on model differences and future directions are provided throughout.
Collapse
Affiliation(s)
- Michael A. Colman
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Blas Echebarria
- Departament de Fisica, Universitat Politècnica de Catalunya-BarcelonaTech, Barcelona, Spain
| | - Daisuke Sato
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Henry Sutanto
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
23
|
Louch WE, Perdreau-Dahl H, Edwards AG. Image-Driven Modeling of Nanoscopic Cardiac Function: Where Have We Come From, and Where Are We Going? Front Physiol 2022; 13:834211. [PMID: 35356084 PMCID: PMC8959215 DOI: 10.3389/fphys.2022.834211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Complementary developments in microscopy and mathematical modeling have been critical to our understanding of cardiac excitation-contraction coupling. Historically, limitations imposed by the spatial or temporal resolution of imaging methods have been addressed through careful mathematical interrogation. Similarly, limitations imposed by computational power have been addressed by imaging macroscopic function in large subcellular domains or in whole myocytes. As both imaging resolution and computational tractability have improved, the two approaches have nearly merged in terms of the scales that they can each be used to interrogate. With this review we will provide an overview of these advances and their contribution to understanding ventricular myocyte function, including exciting developments over the last decade. We specifically focus on experimental methods that have pushed back limits of either spatial or temporal resolution of nanoscale imaging (e.g., DNA-PAINT), or have permitted high resolution imaging on large cellular volumes (e.g., serial scanning electron microscopy). We also review the progression of computational approaches used to integrate and interrogate these new experimental data sources, and comment on near-term advances that may unify understanding of the underlying biology. Finally, we comment on several outstanding questions in cardiac physiology that stand to benefit from a concerted and complementary application of these new experimental and computational methods.
Collapse
Affiliation(s)
- William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Harmonie Perdreau-Dahl
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | | |
Collapse
|
24
|
Abstract
In mammalian cardiac myocytes, the plasma membrane includes the surface sarcolemma but also a network of membrane invaginations called transverse (t-) tubules. These structures carry the action potential deep into the cell interior, allowing efficient triggering of Ca2+ release and initiation of contraction. Once thought to serve as rather static enablers of excitation-contraction coupling, recent work has provided a newfound appreciation of the plasticity of the t-tubule network's structure and function. Indeed, t-tubules are now understood to support dynamic regulation of the heartbeat across a range of timescales, during all stages of life, in both health and disease. This review article aims to summarize these concepts, with consideration given to emerging t-tubule regulators and their targeting in future therapies.
Collapse
Affiliation(s)
- Katharine M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo Norway
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
25
|
Abstract
Junctophilins (JPHs) comprise a family of structural proteins that connect the plasma membrane to intracellular organelles such as the endo/sarcoplasmic reticulum. Tethering of these membrane structures results in the formation of highly organized subcellular junctions that play important signaling roles in all excitable cell types. There are four JPH isoforms, expressed primarily in muscle and neuronal cell types. Each JPH protein consists of 6 'membrane occupation and recognition nexus' (MORN) motifs, a joining region connecting these to another set of 2 MORN motifs, a putative alpha-helical region, a divergent region exhibiting low homology between JPH isoforms, and a carboxy-terminal transmembrane region anchoring into the ER/SR membrane. JPH isoforms play essential roles in developing and maintaining subcellular membrane junctions. Conversely, inherited mutations in JPH2 cause hypertrophic or dilated cardiomyopathy, while trinucleotide expansions in the JPH3 gene cause Huntington Disease-Like 2. Loss of JPH1 protein levels can cause skeletal myopathy, while loss of cardiac JPH2 levels causes heart failure and atrial fibrillation, among other disease. This review will provide a comprehensive overview of the JPH gene family, phylogeny, and evolutionary analysis of JPH genes and other MORN domain proteins. JPH biogenesis, membrane tethering, and binding partners will be discussed, as well as functional roles of JPH isoforms in excitable cells. Finally, potential roles of JPH isoform deficits in human disease pathogenesis will be reviewed.
Collapse
Affiliation(s)
- Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Department of Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States; Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Pediatrics (Cardiology), Neuroscience, and Center for Space Medicine, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
26
|
Wegener JW, Wagdi A, Wagner E, Katschinski DM, Hasenfuss G, Bruegmann T, Lehnart SE. The RyR2-R2474S Mutation Sensitizes Cardiomyocytes and Hearts to Catecholaminergic Stress-Induced Oxidation of the Mitochondrial Glutathione Pool. Front Physiol 2021; 12:777770. [PMID: 34955889 PMCID: PMC8696262 DOI: 10.3389/fphys.2021.777770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/19/2021] [Indexed: 11/24/2022] Open
Abstract
Missense mutations in the cardiac ryanodine receptor type 2 (RyR2) characteristically cause catecholaminergic arrhythmias. Reminiscent of the phenotype in patients, RyR2-R2474S knockin mice develop exercise-induced ventricular tachyarrhythmias. In cardiomyocytes, increased mitochondrial matrix Ca2+ uptake was recently linked to non-linearly enhanced ATP synthesis with important implications for cardiac redox metabolism. We hypothesize that catecholaminergic stimulation and contractile activity amplify mitochondrial oxidation pathologically in RyR2-R2474S cardiomyocytes. To investigate this question, we generated double transgenic RyR2-R2474S mice expressing a mitochondria-restricted fluorescent biosensor to monitor the glutathione redox potential (EGSH). Electrical field pacing-evoked RyR2-WT and RyR2-R2474S cardiomyocyte contractions resulted in a small but significant baseline EGSH increase. Importantly, β-adrenergic stimulation resulted in excessive EGSH oxidization of the mitochondrial matrix in RyR2-R2474S cardiomyocytes compared to baseline and RyR2-WT control. Physiologically β-adrenergic stimulation significantly increased mitochondrial EGSH further in intact beating RyR2-R2474S but not in RyR2-WT control Langendorff perfused hearts. Finally, this catecholaminergic EGSH increase was significantly attenuated following treatment with the RyR2 channel blocker dantrolene. Together, catecholaminergic stimulation and increased diastolic Ca2+ leak induce a strong, but dantrolene-inhibited mitochondrial EGSH oxidization in RyR2-R2474S cardiomyocytes.
Collapse
Affiliation(s)
- Jörg W Wegener
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), Georg-August University of Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Ahmed Wagdi
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany
| | - Eva Wagner
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany
| | - Dörthe M Katschinski
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Tobias Bruegmann
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), Georg-August University of Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany
| | - Stephan E Lehnart
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University of Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), Georg-August University of Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
27
|
Hoang-Trong TM, Ullah A, Lederer WJ, Jafri MS. A Stochastic Spatiotemporal Model of Rat Ventricular Myocyte Calcium Dynamics Demonstrated Necessary Features for Calcium Wave Propagation. MEMBRANES 2021; 11:989. [PMID: 34940490 PMCID: PMC8706945 DOI: 10.3390/membranes11120989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022]
Abstract
Calcium (Ca2+) plays a central role in the excitation and contraction of cardiac myocytes. Experiments have indicated that calcium release is stochastic and regulated locally suggesting the possibility of spatially heterogeneous calcium levels in the cells. This spatial heterogeneity might be important in mediating different signaling pathways. During more than 50 years of computational cell biology, the computational models have been advanced to incorporate more ionic currents, going from deterministic models to stochastic models. While periodic increases in cytoplasmic Ca2+ concentration drive cardiac contraction, aberrant Ca2+ release can underly cardiac arrhythmia. However, the study of the spatial role of calcium ions has been limited due to the computational expense of using a three-dimensional stochastic computational model. In this paper, we introduce a three-dimensional stochastic computational model for rat ventricular myocytes at the whole-cell level that incorporate detailed calcium dynamics, with (1) non-uniform release site placement, (2) non-uniform membrane ionic currents and membrane buffers, (3) stochastic calcium-leak dynamics and (4) non-junctional or rogue ryanodine receptors. The model simulates spark-induced spark activation and spark-induced Ca2+ wave initiation and propagation that occur under conditions of calcium overload at the closed-cell condition, but not when Ca2+ levels are normal. This is considered important since the presence of Ca2+ waves contribute to the activation of arrhythmogenic currents.
Collapse
Affiliation(s)
- Tuan Minh Hoang-Trong
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (T.M.H.-T.); (A.U.)
| | - Aman Ullah
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (T.M.H.-T.); (A.U.)
| | - William Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Mohsin Saleet Jafri
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; (T.M.H.-T.); (A.U.)
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
28
|
Hoang-Trong MT, Ullah A, Lederer WJ, Jafri MS. Cardiac Alternans Occurs through the Synergy of Voltage- and Calcium-Dependent Mechanisms. MEMBRANES 2021; 11:794. [PMID: 34677560 PMCID: PMC8539281 DOI: 10.3390/membranes11100794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 11/24/2022]
Abstract
Cardiac alternans is characterized by alternating weak and strong beats of the heart. This signaling at the cellular level may appear as alternating long and short action potentials (APs) that occur in synchrony with alternating large and small calcium transients, respectively. Previous studies have suggested that alternans manifests itself through either a voltage dependent mechanism based upon action potential restitution or as a calcium dependent mechanism based on refractoriness of calcium release. We use a novel model of cardiac excitation-contraction (EC) coupling in the rat ventricular myocyte that includes 20,000 calcium release units (CRU) each with 49 ryanodine receptors (RyR2s) and 7 L-type calcium channels that are all stochastically gated. The model suggests that at the cellular level in the case of alternans produced by rapid pacing, the mechanism requires a synergy of voltage- and calcium-dependent mechanisms. The rapid pacing reduces AP duration and magnitude reducing the number of L-type calcium channels activating individual CRUs during each AP and thus increases the population of CRUs that can be recruited stochastically. Elevated myoplasmic and sarcoplasmic reticulum (SR) calcium, [Ca2+]myo and [Ca2+]SR respectively, increases ryanodine receptor open probability (Po) according to our model used in this simulation and this increased the probability of activating additional CRUs. A CRU that opens in one beat is less likely to open the subsequent beat due to refractoriness caused by incomplete refilling of the junctional sarcoplasmic reticulum (jSR). Furthermore, the model includes estimates of changes in Na+ fluxes and [Na+]i and thus provides insight into how changes in electrical activity, [Na+]i and sodium-calcium exchanger activity can modulate alternans. The model thus tracks critical elements that can account for rate-dependent changes in [Na+]i and [Ca2+]myo and how they contribute to the generation of Ca2+ signaling alternans in the heart.
Collapse
Affiliation(s)
- Minh Tuan Hoang-Trong
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (M.T.H.-T.); (A.U.)
| | - Aman Ullah
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (M.T.H.-T.); (A.U.)
| | - William Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Mohsin Saleet Jafri
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (M.T.H.-T.); (A.U.)
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
29
|
Calpain-2 specifically cleaves Junctophilin-2 at the same site as Calpain-1 but with less efficacy. Biochem J 2021; 478:3539-3553. [PMID: 34524407 PMCID: PMC8589432 DOI: 10.1042/bcj20210629] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022]
Abstract
Calpain proteolysis contributes to the pathogenesis of heart failure but the calpain isoforms responsible and their substrate specificities have not been rigorously defined. One substrate, Junctophilin-2 (JP2), is essential for maintaining junctional cardiac dyads and excitation-contraction coupling. We previously demonstrated that mouse JP2 is cleaved by calpain-1 (CAPN1) between Arginine 565 (R565) and Threonine 566 (T566). Recently, calpain-2 (CAPN2) was reported to cleave JP2 at a novel site between Glycine 482 (G482) and Threonine 483 (T483). We aimed to directly compare the contributions of each calpain isoform, their Ca2+ sensitivity, and their cleavage site selection for JP2. We find CAPN1, CAPN2 and their requisite CAPNS1 regulatory subunit are induced by pressure overload stress that is concurrent with JP2 cleavage. Using in vitro calpain cleavage assays, we demonstrate that CAPN1 and CAPN2 cleave JP2 into similar 75 kD N-terminal (JP2NT) and 25 kD C-terminal fragments (JP2CT) with CAPNS1 co-expression enhancing proteolysis. Deletion mutagenesis shows both CAPN1 and CAPN2 require R565/T566 but not G482/T483. When heterologously expressed, the JP2CT peptide corresponding to R565/T566 cleavage approximates the 25 kD species found during cardiac stress while the C-terminal peptide from potential cleavage at G482/T483 produces a 35 kD product. Similar results were obtained for human JP2. Finally, we show that CAPN1 has higher Ca2+ sensitivity and cleavage efficacy than CAPN2 on JP2 and other cardiac substrates including cTnT, cTnI and β2-spectrin. We conclude that CAPN2 cleaves JP2 at the same functionally conserved R565/T566 site as CAPN1 but with less efficacy and suggest heart failure may be targeted through specific inhibition of CAPN1.
Collapse
|
30
|
Benitah JP, Perrier R, Mercadier JJ, Pereira L, Gómez AM. RyR2 and Calcium Release in Heart Failure. Front Physiol 2021; 12:734210. [PMID: 34690808 PMCID: PMC8533677 DOI: 10.3389/fphys.2021.734210] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Heart Failure (HF) is defined as the inability of the heart to efficiently pump out enough blood to maintain the body's needs, first at exercise and then also at rest. Alterations in Ca2+ handling contributes to the diminished contraction and relaxation of the failing heart. While most Ca2+ handling protein expression and/or function has been shown to be altered in many models of experimental HF, in this review, we focus in the sarcoplasmic reticulum (SR) Ca2+ release channel, the type 2 ryanodine receptor (RyR2). Various modifications of this channel inducing alterations in its function have been reported. The first was the fact that RyR2 is less responsive to activation by Ca2+ entry through the L-Type calcium channel, which is the functional result of an ultrastructural remodeling of the ventricular cardiomyocyte, with fewer and disorganized transverse (T) tubules. HF is associated with an elevated sympathetic tone and in an oxidant environment. In this line, enhanced RyR2 phosphorylation and oxidation have been shown in human and experimental HF. After several controversies, it is now generally accepted that phosphorylation of RyR2 at the Calmodulin Kinase II site (S2814) is involved in both the depressed contractile function and the enhanced arrhythmic susceptibility of the failing heart. Diminished expression of the FK506 binding protein, FKBP12.6, may also contribute. While these alterations have been mostly studied in the left ventricle of HF with reduced ejection fraction, recent studies are looking at HF with preserved ejection fraction. Moreover, alterations in the RyR2 in HF may also contribute to supraventricular defects associated with HF such as sinus node dysfunction and atrial fibrillation.
Collapse
Affiliation(s)
| | | | | | | | - Ana M. Gómez
- Signaling and Cardiovascular Pathophysiology—UMR-S 1180, INSERM, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
31
|
Kaplan AD, Joca HC, Boyman L, Greiser M. Calcium Signaling Silencing in Atrial Fibrillation: Implications for Atrial Sodium Homeostasis. Int J Mol Sci 2021; 22:10513. [PMID: 34638854 PMCID: PMC8508839 DOI: 10.3390/ijms221910513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia, affecting more than 33 million people worldwide. Despite important advances in therapy, AF's incidence remains high, and treatment often results in recurrence of the arrhythmia. A better understanding of the cellular and molecular changes that (1) trigger AF and (2) occur after the onset of AF will help to identify novel therapeutic targets. Over the past 20 years, a large body of research has shown that intracellular Ca2+ handling is dramatically altered in AF. While some of these changes are arrhythmogenic, other changes counteract cellular arrhythmogenic mechanisms (Calcium Signaling Silencing). The intracellular Na+ concentration ([Na+])i is a key regulator of intracellular Ca2+ handling in cardiac myocytes. Despite its importance in the regulation of intracellular Ca2+ handling, little is known about [Na+]i, its regulation, and how it might be changed in AF. Previous work suggests that there might be increases in the late component of the atrial Na+ current (INa,L) in AF, suggesting that [Na+]i levels might be high in AF. Indeed, a pharmacological blockade of INa,L has been suggested as a treatment for AF. Here, we review calcium signaling silencing and changes in intracellular Na+ homeostasis during AF. We summarize the proposed arrhythmogenic mechanisms associated with increases in INa,L during AF and discuss the evidence from clinical trials that have tested the pharmacological INa,L blocker ranolazine in the treatment of AF.
Collapse
Affiliation(s)
- Aaron D. Kaplan
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Humberto C. Joca
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| | - Liron Boyman
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| | - Maura Greiser
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| |
Collapse
|
32
|
Setterberg IE, Le C, Frisk M, Li J, Louch WE. The Physiology and Pathophysiology of T-Tubules in the Heart. Front Physiol 2021; 12:718404. [PMID: 34566684 PMCID: PMC8458775 DOI: 10.3389/fphys.2021.718404] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
In cardiomyocytes, invaginations of the sarcolemmal membrane called t-tubules are critically important for triggering contraction by excitation-contraction (EC) coupling. These structures form functional junctions with the sarcoplasmic reticulum (SR), and thereby enable close contact between L-type Ca2+ channels (LTCCs) and Ryanodine Receptors (RyRs). This arrangement in turn ensures efficient triggering of Ca2+ release, and contraction. While new data indicate that t-tubules are capable of exhibiting compensatory remodeling, they are also widely reported to be structurally and functionally compromised during disease, resulting in disrupted Ca2+ homeostasis, impaired systolic and/or diastolic function, and arrhythmogenesis. This review summarizes these findings, while highlighting an emerging appreciation of the distinct roles of t-tubules in the pathophysiology of heart failure with reduced and preserved ejection fraction (HFrEF and HFpEF). In this context, we review current understanding of the processes underlying t-tubule growth, maintenance, and degradation, underscoring the involvement of a variety of regulatory proteins, including junctophilin-2 (JPH2), amphiphysin-2 (BIN1), caveolin-3 (Cav3), and newer candidate proteins. Upstream regulation of t-tubule structure/function by cardiac workload and specifically ventricular wall stress is also discussed, alongside perspectives for novel strategies which may therapeutically target these mechanisms.
Collapse
Affiliation(s)
- Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Christopher Le
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
33
|
Romer SH, Metzger S, Peraza K, Wright MC, Jobe DS, Song LS, Rich MM, Foy BD, Talmadge RJ, Voss AA. A mouse model of Huntington's disease shows altered ultrastructure of transverse tubules in skeletal muscle fibers. J Gen Physiol 2021; 153:211860. [PMID: 33683318 PMCID: PMC7931643 DOI: 10.1085/jgp.202012637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/05/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Huntington’s disease (HD) is a fatal and progressive condition with severe debilitating motor defects and muscle weakness. Although classically recognized as a neurodegenerative disorder, there is increasing evidence of cell autonomous toxicity in skeletal muscle. We recently demonstrated that skeletal muscle fibers from the R6/2 model mouse of HD have a decrease in specific membrane capacitance, suggesting a loss of transverse tubule (t-tubule) membrane in R6/2 muscle. A previous report also indicated that Cav1.1 current was reduced in R6/2 skeletal muscle, suggesting defects in excitation–contraction (EC) coupling. Thus, we hypothesized that a loss and/or disruption of the skeletal muscle t-tubule system contributes to changes in EC coupling in R6/2 skeletal muscle. We used live-cell imaging with multiphoton confocal microscopy and transmission electron microscopy to assess the t-tubule architecture in late-stage R6/2 muscle and found no significant differences in the t-tubule system density, regularity, or integrity. However, electron microscopy images revealed that the cross-sectional area of t-tubules at the triad were 25% smaller in R6/2 compared with age-matched control skeletal muscle. Computer simulation revealed that the resulting decrease in the R6/2 t-tubule luminal conductance contributed to, but did not fully explain, the reduced R6/2 membrane capacitance. Analyses of bridging integrator-1 (Bin1), which plays a primary role in t-tubule formation, revealed decreased Bin1 protein levels and aberrant splicing of Bin1 mRNA in R6/2 muscle. Additionally, the distance between the t-tubule and sarcoplasmic reticulum was wider in R6/2 compared with control muscle, which was associated with a decrease in junctophilin 1 and 2 mRNA levels. Altogether, these findings can help explain dysregulated EC coupling and motor impairment in Huntington’s disease.
Collapse
Affiliation(s)
- Shannon H Romer
- Department of Biological Sciences, Wright State University, Dayton, OH.,Odyssey Systems, Environmental Health Effects Laboratory, Navy Medical Research Unit, Dayton, Wright-Patterson Air Force Base, Dayton, OH
| | - Sabrina Metzger
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| | - Kristiana Peraza
- Department of Biological Sciences, California State Polytechnic University, Pomona, Pomona, CA
| | - Matthew C Wright
- Department of Biological Sciences, California State Polytechnic University, Pomona, Pomona, CA
| | - D Scott Jobe
- Department of Biological Sciences, Wright State University, Dayton, OH
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Mark M Rich
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH
| | - Brent D Foy
- Department of Physics, Wright State University, Dayton, OH
| | - Robert J Talmadge
- Department of Biological Sciences, California State Polytechnic University, Pomona, Pomona, CA
| | - Andrew A Voss
- Department of Biological Sciences, Wright State University, Dayton, OH
| |
Collapse
|
34
|
Pavlaki N, De Jong KA, Geertz B, Nikolaev VO, Froese A. Cardiac Hypertrophy Changes Compartmentation of cAMP in Non-Raft Membrane Microdomains. Cells 2021; 10:cells10030535. [PMID: 33802377 PMCID: PMC8001844 DOI: 10.3390/cells10030535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/21/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
Abstract
3′,5′-Cyclic adenosine monophosphate (cAMP) is a ubiquitous second messenger which plays critical roles in cardiac function and disease. In adult mouse ventricular myocytes (AMVMs), several distinct functionally relevant microdomains with tightly compartmentalized cAMP signaling have been described. At least two types of microdomains reside in AMVM plasma membrane which are associated with caveolin-rich raft and non-raft sarcolemma, each with distinct cAMP dynamics and their differential regulation by receptors and cAMP degrading enzymes phosphodiesterases (PDEs). However, it is still unclear how cardiac disease such as hypertrophy leading to heart failure affects cAMP signals specifically in the non-raft membrane microdomains. To answer this question, we generated a novel transgenic mouse line expressing a highly sensitive Förster resonance energy transfer (FRET)-based biosensor E1-CAAX targeted to non-lipid raft membrane microdomains of AMVMs and subjected these mice to pressure overload induced cardiac hypertrophy. We could detect specific changes in PDE3-dependent compartmentation of β-adrenergic receptor induced cAMP in non-raft membrane microdomains which were clearly different from those occurring in caveolin-rich sarcolemma. This indicates differential regulation and distinct responses of these membrane microdomains to cardiac remodeling.
Collapse
Affiliation(s)
- Nikoleta Pavlaki
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.P.); (K.A.D.J.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany;
| | - Kirstie A. De Jong
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.P.); (K.A.D.J.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany;
| | - Birgit Geertz
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany;
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.P.); (K.A.D.J.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany;
- Correspondence: ; Tel.: +49-(0)40-7410-51391
| | - Alexander Froese
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.P.); (K.A.D.J.); (A.F.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany;
| |
Collapse
|
35
|
Medvedev R, Sanchez-Alonso JL, Alvarez-Laviada A, Rossi S, Dries E, Schorn T, Abdul-Salam VB, Trayanova N, Wojciak-Stothard B, Miragoli M, Faggian G, Gorelik J. Nanoscale Study of Calcium Handling Remodeling in Right Ventricular Cardiomyocytes Following Pulmonary Hypertension. Hypertension 2020; 77:605-616. [PMID: 33356404 DOI: 10.1161/hypertensionaha.120.14858] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pulmonary hypertension is a complex disorder characterized by pulmonary vascular remodeling and right ventricular hypertrophy, leading to right heart failure. The mechanisms underlying this process are not well understood. We hypothesize that the structural remodeling occurring in the cardiomyocytes of the right ventricle affects the cytosolic Ca2+ handling leading to arrhythmias. After 12 days of monocrotaline-induced pulmonary hypertension in rats, epicardial mapping showed electrical remodeling in both ventricles. In myocytes isolated from the hypertensive rats, a combination of high-speed camera and confocal line-scan documented a prolongation of Ca2+ transients along with a higher local Ca2+-release activity. These Ca2+ transients were less synchronous than in controls, likely due to disorganized transverse-axial tubular system. In fact, following pulmonary hypertension, hypertrophied right ventricular myocytes showed significantly reduced number of transverse tubules and increased number of axial tubules; however, Stimulation Emission Depletion microscopy demonstrated that the colocalization of L-type Ca2+ channels and RyR2 (ryanodine receptor 2) remained unchanged. Finally, Stimulation Emission Depletion microscopy and super-resolution scanning patch-clamp analysis uncovered a decrease in the density of active L-type Ca2+ channels in right ventricular myocytes with an elevated open probability of the T-tubule anchored channels. This may represent a general mechanism of how nanoscale structural changes at the early stage of pulmonary hypertension impact on the development of the end stage failing phenotype in the right ventricle.
Collapse
Affiliation(s)
- Roman Medvedev
- From the Dipartimento di Cardiochirurgia, Università degli Studi di Verona, Ospedale Borgo Trento, Italy (R.M., G.F.).,National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.).,Humanitas Clinical and Research Center, Rozzano, Italy (R.M., T.S., M.M.)
| | - Jose L Sanchez-Alonso
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.)
| | - Anita Alvarez-Laviada
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.)
| | - Stefano Rossi
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Parma, Italy (S.R., M.M.)
| | - Eef Dries
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.).,Lab of Experimental Cardiology, University of Leuven, Belgium (E.D.)
| | - Tilo Schorn
- Humanitas Clinical and Research Center, Rozzano, Italy (R.M., T.S., M.M.)
| | - Vahitha B Abdul-Salam
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.)
| | - Natalia Trayanova
- Department of Biomedical Engineering and Alliance for Cardiovascular Diagnostic and Treatment Innovation; Johns Hopkins University; Baltimore, MD (N.T.)
| | - Beata Wojciak-Stothard
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.)
| | - Michele Miragoli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Parma, Italy (S.R., M.M.)
| | | | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom (R.M., J.L.S.-A., A.A.-L., E.D., V.B.A.S., B.W.-S., J.G.)
| |
Collapse
|
36
|
Lu F, Pu WT. The architecture and function of cardiac dyads. Biophys Rev 2020; 12:1007-1017. [PMID: 32661902 PMCID: PMC7429583 DOI: 10.1007/s12551-020-00729-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 07/03/2020] [Indexed: 12/28/2022] Open
Abstract
Cardiac excitation-contraction (EC) coupling, which links plasma membrane depolarization to activation of cardiomyocyte contraction, occurs at dyads, the nanoscopic microdomains formed by apposition of transverse (T)-tubules and junctional sarcoplasmic reticulum (jSR). In a dyadic junction, EC coupling occurs through Ca2+-induced Ca2+ release. Membrane depolarization opens voltage-gated L-type Ca2+ channels (LTCCs) in the T-tubule. The resulting influx of extracellular Ca2+ into the dyadic cleft opens Ca2+ release channels known as ryanodine receptors (RYRs) in the jSR, leading to the rapid increase in cytosolic Ca2+ that triggers sarcomere contraction. The efficacy of LTCC-RYR communication greatly affects a myriad of downstream intracellular signaling events, and it is controlled by many factors, including T-tubule and jSR structure, spatial distribution of ion channels, and regulatory proteins that closely regulate the activities of channels within dyads. Alterations in dyad architecture and/or channel activity are seen in many types of heart disease. This review will focus on the current knowledge regarding cardiac dyad structure and function, their alterations in heart failure, and new approaches to study the composition and function of dyads.
Collapse
Affiliation(s)
- Fujian Lu
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA, 02138, USA.
| |
Collapse
|
37
|
Structural variability of dyads relates to calcium release in rat ventricular myocytes. Sci Rep 2020; 10:8076. [PMID: 32415205 PMCID: PMC7229197 DOI: 10.1038/s41598-020-64840-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cardiac excitation-contraction coupling relies on dyads, the intracellular calcium synapses of cardiac myocytes, where the plasma membrane contacts sarcoplasmic reticulum and where electrical excitation triggers calcium release. The morphology of dyads and dynamics of local calcium release vary substantially. To better understand the correspondence between the structure and the functionality of dyads, we estimated incidences of structurally different dyads and of kinetically different calcium release sites and tested their responsiveness to experimental myocardial injury in left ventricular myocytes of rats. According to the structure of dyads estimated in random electron microscopic images of myocardial tissue, the dyads were sorted into 'compact' or 'loose' types. The calcium release fluxes, triggered at local calcium release sites in patch-clamped ventricular myocytes and recorded by laser scanning confocal fluorescence microscopy, were decomposed into 'early' and 'late' components. ANOVA tests revealed very high correlation between the relative amplitudes of early and late calcium release flux components and the relative occurrences of compact and loose dyads in the control and in the injured myocardium. This finding ascertained the relationship between the structure of dyads and the functionality of calcium release sites and the responsiveness of calcium release sites to physical load in cardiac myocytes.
Collapse
|
38
|
Drum BM, Yuan C, de la Mata A, Grainger N, Santana LF. Junctional sarcoplasmic reticulum motility in adult mouse ventricular myocytes. Am J Physiol Cell Physiol 2020; 318:C598-C604. [PMID: 31967858 DOI: 10.1152/ajpcell.00573.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Excitation-contraction (EC) coupling is the coordinated process by which an action potential triggers cardiac myocyte contraction. EC coupling is initiated in dyads where the junctional sarcoplasmic reticulum (jSR) is in tight proximity to the sarcolemma of cardiac myocytes. Existing models of EC coupling critically depend on dyad stability to ensure the fidelity and strength of EC coupling, where even small variations in ryanodine receptor channel and voltage-gated calcium channel-α 1.2 subunit separation dramatically alter EC coupling. However, dyadic motility has never been studied. Here, we developed a novel strategy to track specific jSR units in dissociated adult ventricular myocytes using photoactivatable fluorescent proteins. We found that the jSR is not static. Instead, we observed dynamic formation and dissolution of multiple dyadic junctions regulated by the microtubule-associated molecular motors kinesin-1 and dynein. Our data support a model where reproducibility of EC coupling results from the activation of a temporally averaged number of SR Ca2+ release units forming and dissolving SR-sarcolemmal junctions. These findings challenge the long-held view that the jSR is an immobile structure and provide insights into the mechanisms underlying its motility.
Collapse
Affiliation(s)
- Benjamin M Drum
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Can Yuan
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Ana de la Mata
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - Nathan Grainger
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, California
| |
Collapse
|
39
|
Vermij SH, Abriel H, Kucera JP. Modeling Depolarization Delay, Sodium Currents, and Electrical Potentials in Cardiac Transverse Tubules. Front Physiol 2020; 10:1487. [PMID: 31920695 PMCID: PMC6916517 DOI: 10.3389/fphys.2019.01487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/21/2019] [Indexed: 01/22/2023] Open
Abstract
T-tubules are invaginations of the lateral membrane of striated muscle cells that provide a large surface for ion channels and signaling proteins, thereby supporting excitation–contraction coupling. T-tubules are often remodeled in heart failure. To better understand the electrical behavior of T-tubules of cardiac cells in health and disease, this study addresses two largely unanswered questions regarding their electrical properties: (1) the delay of T-tubular membrane depolarization and (2) the effects of T-tubular sodium current on T-tubular potentials. Here, we present an elementary computational model to determine the delay in depolarization of deep T-tubular membrane segments as the narrow T-tubular lumen provides resistance against the extracellular current. We compare healthy tubules to tubules with constrictions and diseased tubules from mouse and human, and conclude that constrictions greatly delay T-tubular depolarization, while diseased T-tubules depolarize faster than healthy ones due to tubule widening. Increasing the tubule length non-linearly delays the depolarization. We moreover model the effect of T-tubular sodium current on intraluminal T-tubular potentials. We observe that extracellular potentials become negative during the sodium current transient (up to −40 mV in constricted T-tubules), which feedbacks on sodium channel function (self-attenuation) in a manner resembling ephaptic effects that have been described for intercalated discs where opposing membranes are very close together. The intraluminal potential and sodium current self-attenuation however greatly depend on sodium current conductance. These results show that (1) the changes in passive electrical properties of remodeled T-tubules cannot explain the excitation–contraction coupling defects in diseased cells; and (2) the sodium current may modulate intraluminal potentials. Such extracellular potentials might also affect excitation–contraction coupling and macroscopic conduction.
Collapse
Affiliation(s)
- Sarah Helena Vermij
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | | |
Collapse
|
40
|
Shiferaw Y, Aistrup GL, Louch WE, Wasserstrom JA. Remodeling Promotes Proarrhythmic Disruption of Calcium Homeostasis in Failing Atrial Myocytes. Biophys J 2019; 118:476-491. [PMID: 31889516 DOI: 10.1016/j.bpj.2019.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/19/2019] [Accepted: 12/09/2019] [Indexed: 01/31/2023] Open
Abstract
It is well known that heart failure (HF) typically coexists with atrial fibrillation (AF). However, until now, no clear mechanism has been established that relates HF to AF. In this study, we apply a multiscale computational framework to establish a mechanistic link between atrial myocyte structural remodeling in HF and AF. Using a spatially distributed model of calcium (Ca) signaling, we show that disruption of the spatial relationship between L-type Ca channels (LCCs) and ryanodine receptors results in markedly increased Ca content of the sarcoplasmic reticulum (SR). This increase in SR load is due to changes in the balance between Ca entry via LCCs and Ca extrusion due to the sodium-calcium exchanger after an altered spatial relationship between these signaling proteins. Next, we show that the increased SR load in atrial myocytes predisposes these cells to subcellular Ca waves that occur during the action potential (AP) and are triggered by LCC openings. These waves are common in atrial cells because of the absence of a well-developed t-tubule system in most of these cells. This distinct spatial architecture allows for the presence of a large pool of orphaned ryanodine receptors, which can fire and sustain Ca waves during the AP. Finally, we incorporate our atrial cell model in two-dimensional tissue simulations and demonstrate that triggered wave generation in cells leads to electrical waves in tissue that tend to fractionate to form wavelets of excitation. This fractionation is driven by the underlying stochasticity of subcellular Ca waves, which perturbs AP repolarization and consequently induces localized conduction block in tissue. We outline the mechanism for this effect and argue that it may explain the propensity for atrial arrhythmias in HF.
Collapse
Affiliation(s)
- Yohannes Shiferaw
- Department of Physics, California State University, Northridge, California.
| | - Gary L Aistrup
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J A Wasserstrom
- Department of Medicine (Cardiology) and The Feinberg Cardiovascular and Renal Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
41
|
Chan BYH, Roczkowsky A, Cho WJ, Poirier M, Lee TYT, Mahmud Z, Schulz R. Junctophilin-2 is a target of matrix metalloproteinase-2 in myocardial ischemia-reperfusion injury. Basic Res Cardiol 2019; 114:42. [PMID: 31506724 DOI: 10.1007/s00395-019-0749-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/02/2019] [Indexed: 11/30/2022]
Abstract
Junctophilin-2 is a structural membrane protein that tethers T-tubules to the sarcoplasmic reticulum to allow for coordinated calcium-induced calcium release in cardiomyocytes. Defective excitation-contraction coupling in myocardial ischemia-reperfusion (IR) injury is associated with junctophilin-2 proteolysis. However, it remains unclear whether preventing junctophilin-2 proteolysis improves the recovery of cardiac contractile dysfunction in IR injury. Matrix metalloproteinase-2 (MMP-2) is a zinc and calcium-dependent protease that is activated by oxidative stress in myocardial IR injury and cleaves both intracellular and extracellular substrates. To determine whether junctophilin-2 is targeted by MMP-2, isolated rat hearts were perfused in working mode aerobically or subjected to IR injury with the selective MMP inhibitor ARP-100. IR injury impaired the recovery of cardiac contractile function which was associated with increased degradation of junctophilin-2 and damaged cardiac dyads. In IR hearts, ARP-100 improved the recovery of cardiac contractile function, attenuated junctophilin-2 proteolysis, and prevented ultrastructural damage to the dyad. MMP-2 was co-localized with junctophilin-2 in aerobic and IR hearts by immunoprecipitation and immunohistochemistry. In situ zymography showed that MMP activity was localized to the Z-disc and sarcomere in aerobic hearts and accumulated at sites where the striated JPH-2 staining was disrupted in IR hearts. In vitro proteolysis assays determined that junctophilin-2 is susceptible to proteolysis by MMP-2 and in silico analysis predicted multiple MMP-2 cleavage sites between the membrane occupation and recognition nexus repeats and within the divergent region of junctophilin-2. Degradation of junctophilin-2 by MMP-2 is an early consequence of myocardial IR injury which may initiate a cascade of sequelae leading to impaired contractile function.
Collapse
Affiliation(s)
- Brandon Y H Chan
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Andrej Roczkowsky
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Woo Jung Cho
- Faculty of Medicine and Dentistry Cell Imaging Centre, University of Alberta, Edmonton, AB, Canada
| | - Mathieu Poirier
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Tim Y T Lee
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Zabed Mahmud
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute, 462 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
42
|
Nader M. The SLMAP/Striatin complex: An emerging regulator of normal and abnormal cardiac excitation-contraction coupling. Eur J Pharmacol 2019; 858:172491. [PMID: 31233748 DOI: 10.1016/j.ejphar.2019.172491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/01/2022]
Abstract
The excitation-contraction (E-C) module involves a harmonized correspondence between the sarcolemma and the sarcoplasmic reticulum. This is provided by membrane proteins, which primarily shape the caveolae, the T-tubule/Sarcoplasmic reticulum (TT/SR) junction, and the intercalated discs (ICDs). Distortion of either one of these structures impairs myocardial contraction, and subsequently translates into cardiac failure. Thus, detailed studies on the molecular cues of the E-C module are becoming increasingly necessary to pharmacologically eradicate cardiac failure Herein we reviewed the organization of caveolae, TT/SR junctions, and the ICDs in the heart, with special attention to the Sarcolemma Membrane Associated Protein (SLMAP) and striatin (STRN) in cardiac membranes biology and cardiomyocyte contraction. We emphasized on their in vivo and in vitro signaling in cardiac function/dysfunction. SLMAP is a cardiac membrane protein that plays an important role in E-C coupling and the adrenergic response of the heart. Similarly, STRN is a dynamic protein that is also involved in cardiac E-C coupling and ICD-related cardiomyopathies. Both SLMAP and STRN are linked to cardiac conditions, including heart failure, and their role in cardiomyocyte function was elucidated in our laboratory. They interact together in a protein complex that holds therapeutic potentials for cardiac dysfunction. This review is the first of its kind to conceptualize the role of the SLMAP/STRN complex in cardiac function and failure. It provides in depth information on the signaling of these two proteins and projects their interaction as a novel therapeutic target for cardiac failure.
Collapse
Affiliation(s)
- Moni Nader
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, 11533, P.O. Box 50927, Saudi Arabia; Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
43
|
Saeki T, Suzuki Y, Yamamura H, Takeshima H, Imaizumi Y. A junctophilin-caveolin interaction enables efficient coupling between ryanodine receptors and BK Ca channels in the Ca 2+ microdomain of vascular smooth muscle. J Biol Chem 2019; 294:13093-13105. [PMID: 31308177 PMCID: PMC6721949 DOI: 10.1074/jbc.ra119.008342] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/09/2019] [Indexed: 01/09/2023] Open
Abstract
Functional coupling between large-conductance Ca2+-activated K+ (BKCa) channels in the plasma membrane (PM) and ryanodine receptors (RyRs) in the sarcoplasmic reticulum (SR) is an essential mechanism for regulating mechanical force in most smooth muscle (SM) tissues. Spontaneous Ca2+ release through RyRs (Ca2+ sparks) and subsequent BKCa channel activation occur within the PM-SR junctional sites. We report here that a molecular interaction of caveolin-1 (Cav1), a caveola-forming protein, with junctophilin-2 (JP2), a bridging protein between PM and SR, positions BKCa channels near RyRs in SM cells (SMCs) and thereby contributes to the formation of a molecular complex essential for Ca2+ microdomain function. Approximately half of all Ca2+ sparks occurred within a close distance (<400 nm) from fluorescently labeled JP2 or Cav1 particles, when they were moderately expressed in primary SMCs from mouse mesenteric artery. The removal of caveolae by genetic Cav1 ablation or methyl-β-cyclodextrin treatments significantly reduced coupling efficiency between Ca2+ sparks and BKCa channel activity in SMCs, an effect also observed after JP2 knockdown in SMCs. A 20-amino acid-long region in JP2 appeared to be essential for the observed JP2-Cav1 interaction, and we also observed an interaction between JP2 and the BKCa channel. It can be concluded that the JP2-Cav1 interaction provides a structural and functional basis for the Ca2+ microdomain at PM-SR junctions and mediates cross-talk between RyRs and BKCa channels, converts local Ca2+ sparks into membrane hyperpolarization, and contributes to stabilizing resting tone in SMCs.
Collapse
Affiliation(s)
- Takanori Saeki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| |
Collapse
|
44
|
A fundamental evaluation of the electrical properties and function of cardiac transverse tubules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118502. [PMID: 31269418 DOI: 10.1016/j.bbamcr.2019.06.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/07/2019] [Accepted: 06/28/2019] [Indexed: 11/20/2022]
Abstract
This work discusses active and passive electrical properties of transverse (T-)tubules in ventricular cardiomyocytes to understand the physiological roles of T-tubules. T-tubules are invaginations of the lateral membrane that provide a large surface for calcium-handling proteins to facilitate sarcomere shortening. Higher heart rates correlate with higher T-tubular densities in mammalian ventricular cardiomyocytes. We assess ion dynamics in T-tubules and the effects of sodium current in T-tubules on the extracellular potential, which leads to a partial reduction of the sodium current in deep segments of a T-tubule. We moreover reflect on the impact of T-tubules on macroscopic conduction velocity, integrating fundamental principles of action potential propagation and conduction. We also theoretically assess how the conduction velocity is affected by different T-tubular sodium current densities. Lastly, we critically assess literature on ion channel expression to determine whether action potentials can be initiated in T-tubules.
Collapse
|
45
|
Liu C, Spinozzi S, Chen JY, Fang X, Feng W, Perkins G, Cattaneo P, Guimarães-Camboa N, Dalton ND, Peterson KL, Wu T, Ouyang K, Fu XD, Evans SM, Chen J. Nexilin Is a New Component of Junctional Membrane Complexes Required for Cardiac T-Tubule Formation. Circulation 2019; 140:55-66. [PMID: 30982350 PMCID: PMC6889818 DOI: 10.1161/circulationaha.119.039751] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/02/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Membrane contact sites are fundamental for transmission and translation of signals in multicellular organisms. The junctional membrane complexes in the cardiac dyads, where transverse (T) tubules are juxtaposed to the sarcoplasmic reticulum, are a prime example. T-tubule uncoupling and remodeling are well-known features of cardiac disease and heart failure. Even subtle alterations in the association between T-tubules and the junctional sarcoplasmic reticulum can cause serious cardiac disorders. NEXN (nexilin) has been identified as an actin-binding protein, and multiple mutations in the NEXN gene are associated with cardiac diseases, but the precise role of NEXN in heart function and disease is still unknown. METHODS Nexn global and cardiomyocyte-specific knockout mice were generated. Comprehensive phenotypic and RNA sequencing and mass spectrometry analyses were performed. Heart tissue samples and isolated single cardiomyocytes were analyzed by electron and confocal microscopy. RESULTS Global and cardiomyocyte-specific loss of Nexn in mice resulted in a rapidly progressive dilated cardiomyopathy. In vivo and in vitro analyses revealed that NEXN interacted with junctional sarcoplasmic reticulum proteins, was essential for optimal calcium transients, and was required for initiation of T-tubule invagination and formation. CONCLUSIONS These results demonstrated that NEXN is a pivotal component of the junctional membrane complex and is required for initiation and formation of T-tubules, thus providing insight into mechanisms underlying cardiomyopathy in patients with mutations in NEXN.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/metabolism
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cell Membrane/genetics
- Cell Membrane/metabolism
- Cell Membrane/pathology
- Cells, Cultured
- Intercellular Junctions/genetics
- Intercellular Junctions/metabolism
- Intercellular Junctions/pathology
- Mice
- Mice, Knockout
- Mice, Transgenic
- Microfilament Proteins/deficiency
- Microfilament Proteins/genetics
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
Collapse
Affiliation(s)
- Canzhao Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Simone Spinozzi
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia-Yu Chen
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Wei Feng
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Paola Cattaneo
- National Research Council, Institute of Genetics and Biomedical Research, Milan 20138, Italy
- Humanitas Clinical and Research Center, Rozzano 20089, Italy
| | - Nuno Guimarães-Camboa
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Berlin 13347, Germany
| | - Nancy D. Dalton
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kirk L. Peterson
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Sylvia M. Evans
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
46
|
Vaidyanathan R, Van Ert H, Haq KT, Morotti S, Esch S, McCune EC, Grandi E, Eckhardt LL. Inward Rectifier Potassium Channels (Kir2.x) and Caveolin-3 Domain-Specific Interaction: Implications for Purkinje Cell-Dependent Ventricular Arrhythmias. Circ Arrhythm Electrophysiol 2019; 11:e005800. [PMID: 29326130 DOI: 10.1161/circep.117.005800] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/30/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND In human cardiac ventricle, IK1 is mainly comprised Kir2.1, but Kir2.2 and Kir2.3 heterotetramers occur and modulate IK1. Long-QT syndrome-9-associated CAV3 mutations cause decreased Kir2.1 current density, but Kir2.x heterotetramers have not been studied. Here, we determine the effect of long-QT syndrome-9-CAV3 mutation F97C on Kir2.x homo- and heterotetramers and model-associated arrhythmia mechanisms. METHODS AND RESULTS Super-resolution microscopy, co-immunoprecipitation, cellular electrophysiology, on-cell Western blotting, and simulation of Purkinje and ventricular myocyte mathematical models were used. Kir2.x isoforms have unique subcellular colocalization in human cardiomyocytes and coimmunoprecipitate with Cav3. F97C-Cav3 decreased peak inward Kir2.2 current density by 50% (-120 mV; P=0.019) and peak outward by 75% (-40 mV; P<0.05) but did not affect Kir2.3 current density. FRET (Förster resonance energy transfer) efficiency for Kir2.2 with Cav3 is high, and on-cell Western blotting demonstrates decreased Kir2.2 membrane expression with F97C-Cav3. Cav3-F97C reduced peak inward and outward current density of Kir2.2/Kir2.1 or Kir2.2/Kir2.3 heterotetramers (P<0.05). Only Cav3 scaffolding and membrane domains co-immunoprecipitation with Kir2.1 and Kir2.2 and Kir2.x-N-terminal Cav3 binding motifs are required for interaction. Mathematical Purkinje, but not ventricular, myocyte model incorporating simulated current reductions, predicts spontaneous delayed after-depolarization-mediated triggered activity. CONCLUSIONS Kir2.x isoforms have a unique intracellular pattern of distribution in association with specific Cav3 domains and that critically depends on interaction with N-terminal Kir2.x Cav3-binding motifs. Long-QT syndrome-9-CAV3 mutation differentially regulates current density and cell surface expression of Kir2.x homomeric and heteromeric channels. Mathematical Purkinje cell model incorporating experimental findings suggests delayed after-depolarization-type triggered activity as a possible arrhythmia mechanism.
Collapse
Affiliation(s)
- Ravi Vaidyanathan
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Hanora Van Ert
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Kazi T Haq
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Stefano Morotti
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Samuel Esch
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Elise C McCune
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Eleonora Grandi
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.)
| | - Lee L Eckhardt
- From the Division of Cardiovascular Medicine and the Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin, Madison (R.V., H.V.E., S.E., E.C.M., L.L.E.); and Department of Pharmacology, University of California Davis (K.T.H., S.M., E.G.).
| |
Collapse
|
47
|
Brandenburg S, Pawlowitz J, Eikenbusch B, Peper J, Kohl T, Mitronova GY, Sossalla S, Hasenfuss G, Wehrens XH, Kohl P, Rog-Zielinska EA, Lehnart SE. Junctophilin-2 expression rescues atrial dysfunction through polyadic junctional membrane complex biogenesis. JCI Insight 2019; 4:127116. [PMID: 31217359 DOI: 10.1172/jci.insight.127116] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/16/2019] [Indexed: 12/28/2022] Open
Abstract
Atrial dysfunction is highly prevalent and associated with increased severity of heart failure. While rapid excitation-contraction coupling depends on axial junctions in atrial myocytes, the molecular basis of atrial loss of function remains unclear. We identified approximately 5-fold lower junctophilin-2 levels in atrial compared with ventricular tissue in mouse and human hearts. In atrial myocytes, this resulted in subcellular expression of large junctophilin-2 clusters at axial junctions, together with highly phosphorylated ryanodine receptor (RyR2) channels. To investigate the contribution of junctophilin-2 to atrial pathology in adult hearts, we developed a cardiomyocyte-selective junctophilin-2-knockdown model with 0 mortality. Junctophilin-2 knockdown in mice disrupted atrial RyR2 clustering and contractility without hypertrophy or interstitial fibrosis. In contrast, aortic pressure overload resulted in left atrial hypertrophy with decreased junctophilin-2 and RyR2 expression, disrupted axial junctions, and atrial fibrosis. Whereas pressure overload accrued atrial dysfunction and heart failure with 40% mortality, additional junctophilin-2 knockdown greatly exacerbated atrial dysfunction with 100% mortality. Strikingly, transgenic junctophilin-2 overexpression restored atrial contractility and survival through de novo biogenesis of polyadic junctional membrane complexes maintained after pressure overload. Our data show a central role of junctophilin-2 cluster disruption in atrial hypertrophy and identify transgenic augmentation of junctophilin-2 as a disease-mitigating rationale to improve atrial dysfunction and prevent heart failure deterioration.
Collapse
Affiliation(s)
- Sören Brandenburg
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Jan Pawlowitz
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Benjamin Eikenbusch
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Jonas Peper
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Kohl
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Gyuzel Y Mitronova
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Samuel Sossalla
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerd Hasenfuss
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany
| | - Xander Ht Wehrens
- Cardiovascular Research Institute - Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Kohl
- University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Eva A Rog-Zielinska
- University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stephan E Lehnart
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany.,BioMET, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Chasing Uptake: Super-Resolution Microscopy in Endocytosis and Phagocytosis. Trends Cell Biol 2019; 29:727-739. [PMID: 31227311 DOI: 10.1016/j.tcb.2019.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 11/21/2022]
Abstract
Since their invention about two decades ago, super-resolution microscopes have become a method of choice in cell biology. Owing to a spatial resolution below 50 nm, smaller than the size of most organelles, and an order of magnitude better than the diffraction limit of conventional light microscopes, super-resolution microscopy is a powerful technique for resolving intracellular trafficking. In this review we discuss discoveries in endocytosis and phagocytosis that have been made possible by super-resolution microscopy - from uptake at the plasma membrane, endocytic coat formation, and cytoskeletal rearrangements to endosomal maturation. The detailed visualization of the diverse molecular assemblies that mediate endocytic uptake will provide a better understanding of how cells ingest extracellular material.
Collapse
|
49
|
Alsina KM, Hulsurkar M, Brandenburg S, Kownatzki-Danger D, Lenz C, Urlaub H, Abu-Taha I, Kamler M, Chiang DY, Lahiri SK, Reynolds JO, Quick AP, Scott L, Word TA, Gelves MD, Heck AJR, Li N, Dobrev D, Lehnart SE, Wehrens XHT. Loss of Protein Phosphatase 1 Regulatory Subunit PPP1R3A Promotes Atrial Fibrillation. Circulation 2019; 140:681-693. [PMID: 31185731 DOI: 10.1161/circulationaha.119.039642] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Abnormal calcium (Ca2+) release from the sarcoplasmic reticulum (SR) contributes to the pathogenesis of atrial fibrillation (AF). Increased phosphorylation of 2 proteins essential for normal SR-Ca2+ cycling, the type-2 ryanodine receptor (RyR2) and phospholamban (PLN), enhances the susceptibility to AF, but the underlying mechanisms remain unclear. Protein phosphatase 1 (PP1) limits steady-state phosphorylation of both RyR2 and PLN. Proteomic analysis uncovered a novel PP1-regulatory subunit (PPP1R3A [PP1 regulatory subunit type 3A]) in the RyR2 macromolecular channel complex that has been previously shown to mediate PP1 targeting to PLN. We tested the hypothesis that reduced PPP1R3A levels contribute to AF pathogenesis by reducing PP1 binding to both RyR2 and PLN. METHODS Immunoprecipitation, mass spectrometry, and complexome profiling were performed from the atrial tissue of patients with AF and from cardiac lysates of wild-type and Pln-knockout mice. Ppp1r3a-knockout mice were generated by CRISPR-mediated deletion of exons 2 to 3. Ppp1r3a-knockout mice and wild-type littermates were subjected to in vivo programmed electrical stimulation to determine AF susceptibility. Isolated atrial cardiomyocytes were used for Stimulated Emission Depletion superresolution microscopy and confocal Ca2+ imaging. RESULTS Proteomics identified the PP1-regulatory subunit PPP1R3A as a novel RyR2-binding partner, and coimmunoprecipitation confirmed PPP1R3A binding to RyR2 and PLN. Complexome profiling and Stimulated Emission Depletion imaging revealed that PLN is present in the PPP1R3A-RyR2 interaction, suggesting the existence of a previously unknown SR nanodomain composed of both RyR2 and PLN/sarco/endoplasmic reticulum calcium ATPase-2a macromolecular complexes. This novel RyR2/PLN/sarco/endoplasmic reticulum calcium ATPase-2a complex was also identified in human atria. Genetic ablation of Ppp1r3a in mice impaired binding of PP1 to both RyR2 and PLN. Reduced PP1 targeting was associated with increased phosphorylation of RyR2 and PLN, aberrant SR-Ca2+ release in atrial cardiomyocytes, and enhanced susceptibility to pacing-induced AF. Finally, PPP1R3A was progressively downregulated in the atria of patients with paroxysmal and persistent (chronic) AF. CONCLUSIONS PPP1R3A is a novel PP1-regulatory subunit within the RyR2 channel complex. Reduced PPP1R3A levels impair PP1 targeting and increase phosphorylation of both RyR2 and PLN. PPP1R3A deficiency promotes abnormal SR-Ca2+ release and increases AF susceptibility in mice. Given that PPP1R3A is downregulated in patients with AF, this regulatory subunit may represent a new target for AF therapeutic strategies.
Collapse
Affiliation(s)
- Katherina M Alsina
- Integrative Molecular and Biomedical Sciences (K.M.A., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Mohit Hulsurkar
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Sören Brandenburg
- Cellular Biophysics and Translational Cardiology Research Section, Heart Research Center Göttingen, and Department of Cardiology & Pneumology, University Medical Center of Göttingen, Germany (S.B., D.K.-D., S.E.L.)
| | - Daniel Kownatzki-Danger
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Cellular Biophysics and Translational Cardiology Research Section, Heart Research Center Göttingen, and Department of Cardiology & Pneumology, University Medical Center of Göttingen, Germany (S.B., D.K.-D., S.E.L.)
| | - Christof Lenz
- Institute of Clinical Chemistry, University Medical Center Göttingen, and Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Germany (C.L., H.U.)
| | - Henning Urlaub
- Institute of Clinical Chemistry, University Medical Center Göttingen, and Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Germany (C.L., H.U.)
| | - Issam Abu-Taha
- Institute of Pharmacology, West Germany Heart and Vascular Center (I.A.-T., D.D.), University Duisburg-Essen, Germany
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery Huttrop (M.K.), University Duisburg-Essen, Germany
| | - David Y Chiang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.Y.C.)
| | - Satadru K Lahiri
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Julia O Reynolds
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Ann P Quick
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Larry Scott
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Tarah A Word
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Maria D Gelves
- Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands (A.J.R.H.).,Netherlands Proteomics Centre, Utrecht (A.J.R.H.)
| | - Na Li
- Integrative Molecular and Biomedical Sciences (K.M.A., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Medicine (Cardiology), Baylor College of Medicine (N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Dobromir Dobrev
- Institute of Pharmacology, West Germany Heart and Vascular Center (I.A.-T., D.D.), University Duisburg-Essen, Germany.,DZHK (German Centre for Cardiovascular Research) site Goettingen (S.E.L.)
| | - Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Research Section, Heart Research Center Göttingen, and Department of Cardiology & Pneumology, University Medical Center of Göttingen, Germany (S.B., D.K.-D., S.E.L.)
| | - Xander H T Wehrens
- Integrative Molecular and Biomedical Sciences (K.M.A., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Cardiovascular Research Institute (K.MA., M.H., D.Y.C., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., M.D.G., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (M.H., S.K.L., J.O.R., A.P.Q., L.S., T.A.W., N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Medicine (Cardiology), Baylor College of Medicine (N.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Pediatrics (Cardiology) (X.H.T.W.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
50
|
Lawless M, Caldwell JL, Radcliffe EJ, Smith CER, Madders GWP, Hutchings DC, Woods LS, Church SJ, Unwin RD, Kirkwood GJ, Becker LK, Pearman CM, Taylor RF, Eisner DA, Dibb KM, Trafford AW. Phosphodiesterase 5 inhibition improves contractile function and restores transverse tubule loss and catecholamine responsiveness in heart failure. Sci Rep 2019; 9:6801. [PMID: 31043634 PMCID: PMC6494852 DOI: 10.1038/s41598-019-42592-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is characterized by poor survival, a loss of catecholamine reserve and cellular structural remodeling in the form of disorganization and loss of the transverse tubule network. Indeed, survival rates for HF are worse than many common cancers and have not improved over time. Tadalafil is a clinically relevant drug that blocks phosphodiesterase 5 with high specificity and is used to treat erectile dysfunction. Using a sheep model of advanced HF, we show that tadalafil treatment improves contractile function, reverses transverse tubule loss, restores calcium transient amplitude and the heart's response to catecholamines. Accompanying these effects, tadalafil treatment normalized BNP mRNA and prevented development of subjective signs of HF. These effects were independent of changes in myocardial cGMP content and were associated with upregulation of both monomeric and dimerized forms of protein kinase G and of the cGMP hydrolyzing phosphodiesterases 2 and 3. We propose that the molecular switch for the loss of transverse tubules in HF and their restoration following tadalafil treatment involves the BAR domain protein Amphiphysin II (BIN1) and the restoration of catecholamine sensitivity is through reductions in G-protein receptor kinase 2, protein phosphatase 1 and protein phosphatase 2 A abundance following phosphodiesterase 5 inhibition.
Collapse
Affiliation(s)
- Michael Lawless
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Jessica L Caldwell
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Emma J Radcliffe
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Charlotte E R Smith
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - George W P Madders
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - David C Hutchings
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Lori S Woods
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Stephanie J Church
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Richard D Unwin
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Graeme J Kirkwood
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Lorenz K Becker
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Charles M Pearman
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Rebecca F Taylor
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - David A Eisner
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Katharine M Dibb
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom.
| |
Collapse
|